Skip to main content

Full text of "USPTO Patents Application 10564397"

See other formats


Document made available under the 
Patent Cooperation Treaty (PCT) 



International application number: PCT7US04/022605 
International filing date: 14 July 2004 (14.07.2004) 

Document type: Certified copy of priority document 

Document details: Country/Office: US 

Number: 60/486,750 

Filing date: 14 July 2003 (14.07.2003) 



Date of receipt at the International Bureau: 11 November 2004 (11.11.2004) 



Remark: Priority document submitted or transmitted to the International Bureau in 
compliance with Rule 17.1(a) or (b) 




World Intellectual Property Organization (WIPO) - Geneva, Switzerland 
Organisation Mondiale de la Propriete Intellectuelle (OMPI) - Geneve, Suisse 



UNITED STATES DEPARTMENT OF COMMERCE 
United States Patent and Trademark Office 

November 05, 2004 



THIS IS TO CERTIFY THAT ANNEXED HERETO IS A TRUE COPY FROM 
THE RECORDS OF THE UNITED STATES PATENT AND TRADEMARK 
OFFICE OF THOSE PAPERS OF THE BELOW IDENTIFIED PATENT 
APPLICATION THAT MET THE REQUIREMENTS TO BE GRANTED A 
FILING DATE. 



APPLICATION NUMBER: 60/486,750 
FILING DATE: July 14, 2003 

RELATED PCT APPLICATION NUMBER: PCT/US04/22605 



Certified by- 




Jon W Dudas 

Acting Under Secretary of Commerce 
for Intellectual Property 
and Acting Director of the U.S. 
Patent and Trademark Office 



' Please type a plus sign (+) inside this box ** ^ [+| 

Revised PTOySB/1 6 (8-O0) 
Approved for use through 10/31/2002. OMB 0651-0032 
Patent and Trademark Office; U.S. DEPARTMENT OF COMMERCE 
Under the Paperwork Reduction Act of 1395, no persons are required to respond to a collection of information unless it displays a valid OMB control number. 

Attorney Docket No. 41860-189496 

PROVISIONAL APPLICATION FOR PATENT COVER SHEET 
This is a request for filing a PROVISIONAL APPLICATION FOR PATENT under 37 CFR 1.53 (c). 



o 

H 



.to 
r — 



INVENTOR(S) 



Given Name (first and middle [if any]) 



Family Name or Surname 



Residence 
(City and either State or Foreign Country) 



Evans 

Jane 

Simon 
Duncan 
Yoshikazu 

Roger 

John 
Matasuke 



TARACHA 

GLEW 
GRAHAM 
MWANGI 
HONDA 
PELLE 
TONUKARI 
YAMAGE 



Nairobi, 
Nairobi, 
Nairobi, 
Nairobi, 
Nairobi, 
Nairobi, 
Nairobi, 
Nairobi, 



KENYA 
KENYA 
KENYA 
KENYA 
KENYA 
KENYA 
KENYA 
KENYA 



no 
mvo 



D Additional inventors are being named on the separately numbered sheets attached hereto 



TITLE OF THE INVENTION (280 characters max) 



EAST COAST FEVER VACCINE BASED ON CTL-SPECIFIC SCHIZONT ANTIGENS 



Direct all correspondence to: 
E Customer Number 
OR 



26694 



CORRESPONDENCE ADDRESS 
► 



Type Customer Number here 



26694 

PATENT TRADEMARK OFFICE 



Firm or 

Individual Name 



VENABLE 



Address 



P.O. Box 34385 



Address 



City 



Washington 



State 



DC 



ZIP 



20043-9998 



Country 



U.S.A. 



Telepho 



202.962.4800 



Fax 



202.962.8300 



ENCLOSED APPLICATION PARTS (check all that apply) 



^ Specification Number of Pages 
[J Drawing(s) Number of Sheets 



53_ 



!EI Application Data Sheet. See 37 CFR 1.76 



□ CD(s), Number 

E^l Other (specify) 
Annex A, 68 pages , including Figs 1-23 
Annex B, 7 pages, including Figs 1-6 



METHOD OF PAYMENT OF FILING FEES FOR THIS PROVISIONAL APPLICATION FOR PATENT (check one) 

(3 Applicant claims small entity status. See 37 CFR 1.27. 
^ A check or money order is enclosed to cover the filing fees 



IXI The Commissioner is hereby authorized to charge filing 

fees or credit any overpayment to Deposit Account Number: 
□ Payment by credit card. Form PTO-2038 is attached. 



FILING FEE 
AMOUNT {$) 



22-0261 



80.00 



The invention was made by an agency of the United States Government or under a contract with an agency of 
the United States Government. 

E] No. 

□ Yes, the name of the U.S. GovemmJ&r t 




agency and the Government contract number are: . 



Respectfully submitted, 
SIGNATURE. 



TYPED or PRINTED NAME Michael A. Gollin 



Date 



7/14/03 



TELEPHONE (202) 962-4072 

Imble 



REGISTRATION NO. 
(if appropriate) 

Docket Number: 



31,957 



41860-189496 



(41860-189496) 



1 EAST COAST FEVER VACCINE BASED ON CTL-SPECMC SCHIZONT ANTIGENS 
2 

3 BACKGROUND OF THE INVENTION 

4 Theilerioses are a group of disease syndromes affecting cattle, sheep, goats and 

5 domestic buffalo caused by tick-borne haemo-protozoan parasites in the genus Theileria. 

6 The most economically important diseases include Mediterranean fever, East Coast fever 

7 (ECF) and Malignant theileriosis. Mediterranean fever caused by Theileria annulata 

8 occurs in North Africa, southern Europe, Near East, Middle East and many parts of Far 

9 East Asia with a population of 200 million cattle and buffalo at risk. ECF, caused by T. 

10 parva, affects 30 million cattle in eastern, central and southern Africa. Malignant 

1 1 theileriosis caused by 7. lestoquardi affects sheep and goats in southeastern Europe, 

12 North Africa, the Near and Middle East and southern Russia and neighbouring States. 

13 These parasites belong to the same api-complexan group as Plasmodium falciparum, 

14 Toxoplasma gondii, Cytoxauzoon spp, Eimeria spp and Babesia spp, with a life-cycle 

15 having the arthropod and mammalian components in which sexual and asexual stages 

16 develop, respectively. The pathogenic stages of Theileria parasites differ. T. pan/a causes 

17 a lympho-proliferative disorder in which schizont-infected lymphoblasts are responsible 

18 for the pathogenesis of the disease. On the other hand, anaemic disease caused by T. 

19 lestoquardi and T. sergenti is due to piroplasm-infected erythrocytes while both the 

20 schizont and piroplasm of T. annulata are pathogenic resulting in lympho-proliferative and 

21 anaemic syndromes, respectively. 
22 

23 Currently, theilerioses are controlled largely by tick control using acaricides and through 

24 Infection and treatment" vaccination protocols, of animals at risk. Due to cost and 

25 problems of tick resistance and environmental pollution, control of these diseases through 

26 acaricidal destruction of ticks is not sustainable. Vaccination, on the other hand, while 

27 effective presents with certain shortcomings associated with the use of live vaccines. 

28 Owing to the ease with which to transmit T. annulata, infected blood was originally used 

29 to immunise cattle with parasites of low virulence but were still accompanied by clinical 

30 episodes. With the advent of in vitro cultivation of T. annulata (Sharma et al., 1998) and 

31 the development of bulk culture techniques in the 1960s, significant progress was made 

32 in realising a practical immunisation strategy. Currently, passage-attenuated cultures of T. 

33 annulata are routinely used in national vaccination programs in affected countries. By 

34 contrast, similar efforts to immunise cattle against T parva were unsuccessful. This was 

35 attributed to the failure of attenuated 7. parva parasites to induce immunity. In addition, 

36 much higher numbers of T. pa/va-infected cells were required to infect cattle reliably since 

37 the schizonts of T parva transfer at a low frequency and donor cells get rejected before 

38 successful transfer. T. lestoquardi has also been cultivated in vitro and studies have 

39 shown that attenuated parasites can be used to immunise animals with a degree of 

40 success. 
41 

42 Given the unsuccessful attempts to immunise cattle with attenuated T. parva, subsequent 

43 efforts have focussed on the use of virulent parasites with accompanying chemotherapy. 

44 The rationale of this infection and treatment method (ITM) is to allow the infection to 

45 establish and suppress development of patent clinical disease by administering 

46 theileriacidal drugs. Animals thus immunised were found to be protected against the 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



1 



(41860-189496) 



1 homologous parasite. This vaccination strategy has undergone successive refinement 

2 including the use of cryopreserved triturated tick stabilites containing sporozoites (the 

3 parasite stage infective for cattle lymphocytes) to standardise the infection dose, as well 

4 as simultaneous drug administration. Further improvement of this immunisation approach 

5 has involved the identification and use a combination of parasite stocks to broaden the 

6 immunising spectrum of the vaccine against several field T. pan/a parasite populations. 

7 But the use of local parasite stocks to immunise in areas where they have been isolated 

8 is also practised. ITM immunisation against 7. parva has been tested extensively under 

9 laboratory and field conditions and is now deployed in the affected region to control ECF. 
10 

1 1 ITM is a very efficacious vaccine, however, it has a number of practical limitations that 

12 hinder its application as a sustainable control measure against ECF. Being live, it requires 

13 a cold chain, which is impractical in Africa, and also causes clinical disease if drug 

14 application is inadequate and has the potential to introduce new parasite strains in areas 

15 under the vaccination campaign. The cost (US$10-20 per immunisation) this vaccine is 

16 well beyond the poor farmers afflicted by ECF due to the cost of the drugs and the 

17 requirement for a trained veterinarian to administer the vaccine. Because of these 

18 concerns for the ITM vaccine, a great deal of investment has been put in research work to 

19 develop a vaccine that will be sustainable. 
20 

21 Antigens of parasitic protozoans that induce a protective antibody response against the 

22 development of disease, have been identified. For example, the major merozoite surface 

23 protein of Plasmodium species has been shown to be a target of varying degrees of 

24 protective immunity against the asexual blood stages in rodent and human malaria. 

25 Vaccination of mice with purified P230, the major merozoite surface protein of the rodent 

26 malaria Plasmodium yoelii, has resulted in reduced parasitemias in comparison to 

27 controls upon intravenous challenge with a lethal dose of parasitized erythrocytes (Holder 

28 et al. 1981 . Nature 294:361 ). Mice have also been protected against P. yoelii by passive 

29 transfer of a monoclonal antibody (Mab) specific for P230 (Majarian et al. 1984, J, 

30 Immunol. 132:3131) and against (rodent malaria) Plasmodium chabaudi adami challenge 

31 by passive immunization with a Mab specific for the homologous 250-kDa molecule of this 

32 Plasmodium species (Lew et al. 1989. Proc. Natl. Acad. Sci. USA 86:3768). 
33 

34 A 67 kDa glycoprotein (p67) from the surface of the 7". parva sporozoite has been isolated 

35 (U.S. Patent Number 5273744). Cattle recovering from a single infection with T. parva 

36 sporozoites resist homologous challenge. These animals have weak antibody and T cell 

37 responses to p67. However, when repeatedly exposed to sporozoites, high anti-p67 

38 antibodies are detected which neutralize infectivity of sporozoites. Cattle trials with this 

39 candidate vaccine have demonstrated less than optimal protection despite induction of 

40 strong antibody responses using traditional antigen delivery methods. Protection 

41 engendered following a single infection has been shown to be based on the induction of 

42 class I MHC-restricted CD8 + cytotoxic T lymphocytes (CTLs) whose target parasite 

43 antigens would be prime vaccine candidates. These studies, conducted over a period of 

44 20 years, have made the development of an improved sub-unit vaccine against ECF a 

45 reasonable probability. However, it has been impossible to identify the specific pathogen 

46 antigens that could induce a CD8 + cytotoxic T lymphocyte response. Methods using 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



2 



t « 

(41860-189496) 

1 traditional reagents and cell lines have failed to identify particular antigens that will 

2 stimulate this CD8+ lymphocyte pathway. 
3 

4 

5 

6 SUMMARY OF THE INVENTION 
7 

8 The present invention relates to compositions and methods for the identification of 

9 parasite antigens, such as Theileria parva antigens, that trigger antigen-specific cytotoxic 

10 T lymphocyte (CTLs) responses, for inducing immunoprotection against T. parva in 

1 1 bovine species. More particularly, the method steps wherein the stimulation of 

12 responding lymphocytes to cells transfected by cDNAs encoding parasite antigen, is 

13 measured in a high throughput manner, by the release of soluble factors, such as gamma 

14 interferon, using either an antibody-el ispot assay or a bioassay employing endothelial 

15 cells; the use of immortalized skin fibroblast cells from outbred animals, that have 

16 recovered from exposure, as antigen presenting cells, enables antigen identification, 

17 especially, where cloned bovine MHC class I genes are not available for co-transfection 

18 into COS cells; and/or the resolution of the identity of individual antigens from candidate 

19 cDNA pools. 
20 

21 BRIEF DESCRIPTION OF THE DRAWINGS 
22 

23 FIG. 1 is a photograph on an ethidium bromide-stained agarose gel of total T. parva 

24 schizont RNA, poly(A+) RNA , and poly(A-)RNA. 

25 

26 FIG. 2 is a photograph of an autoradiograph of 32P-labellled double stranded cDNA 

27 copied from poly(A+) T. parva, schizont RNA. 

28 

29 FIG. 3 is a graph of Elispot data, indicating the mean spot forming cells/well when COS-7 

30 cells, co-transfected with a bovine MHC Class I gene and 7". parva schizont cDNA pools, 

31 were co-cultured with cytotoxic T lymphocyte cell (CTL) lines, as measured by CTL 

32 gamma interferon secretion. 

33 

34 FIG. 4 is a graph of Elispot data, indicating the mean spot forming cells/well when COS-7 

35 cells, co-transfected with a bovine MHC Class I gene and 7. pan/a schizont cDNAs 

36 consisting of a pool of 10cDNAs, were co-cultured with cytotoxic T lymphocyte cell (CTL) 

37 lines, as measured by CTL gamma interferon secretion. 

38 

39 FIG. 5 is a graph of Elispot data, indicating the mean spot forming cells/well when COS-7 

40 cells, co-transfected with a bovine MHC Class I gene and individual single T. parva 

41 schizont cDNA clones, were co-cultured with cytotoxic T lymphocyte cell (CTL) lines, as 

42 measured by CTL gamma interferon secretion. 

43 

44 FIG. 6 is a graph of Elispot data, indicating the mean spot forming cells/well when COS-7 

45 cells, co-transfected with a bovine MHC Class I gene and an individual single T. parva 

46 schizont cDNA clone, Tp1 , were co-cultured with cytotoxic T lymphocyte cell (CTL) lines, 

47 as measured by CTL gamma interferon secretion. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



3 



(41860-189496) 



1 

2 FIG. 7 is a graph of Elispot data comparing the ability of COS-7 co-transfected cells to 

3 present target antigen to CTLs, to the ability of immortalized skin fibroblast cells (iSF) to 

4 present target antigen to CTLs. 
5 

6 FIG. 8(a) is a graph of Elispot data 9(b) is a graph of bioassay data. Both assays 

7 measure the secretion of gamma interferon by CTLS stimulated with Tp1 . 

8 

9 FIG. 9 is a graph indicating the % lysis of COS-7 cells, co-transfected with a MHC Class I 
10 gene and a cDNAforTpl. 
11 

12 FIG. 10 is the deduced amino acid sequence of Exonuclease III deleted Tp1 cDNA 

13 plasmids. 
14 

15 FIG. 11 is the deduced amino acid sequence of deleted Tp1 clones. 
16 

17 FIG. 12 is a graph of Elispot data indicating mapping of a Tp1 epitope using a synthetic 

18 peptide library. 
19 

20 FIG. 13 is a graph of Elispot data indicating mapping of the minimal length of the HD6 

21 restricted epitope of Tp1 . 

22 

23 FIG. 14 is a comparison of the deduced amino acid sequences of Tp1 from two different 

24 T. parva strains, Muguga and Marikebuni. 

25 
26 
27 

28 FIG. 1 5 is a graph of Elispot data that indicates the response of CD8+ T cells, harvested 

29 from an immune bull to the HD6 restricted epitope of Tp1 . 

30 

31 FIG. 16 is the DNA sequence and deduced protein sequence of Tp1. 

32 

33 FIG. 17 is the DNA sequence and deduced protein sequence of Tp4. 

34 

35 FIG. 18 is the DNA sequence and deduced protein sequence of Tp5. 

36 

37 FIG. 19 is the DNA sequence and deduced protein sequence of Tp7. 

38 

39 FIG. 20 is the DNA sequence and deduced protein sequence of Tp8. 
40 

41 FIG. 21 (a) is a photograph of a Coomassie stained gel and an anti-His-tag immunoblot 

42 (Western blot) of Tp1 expression, using a bacterial expression vector; FIG. 22(b) is a 

43 photograph of a Coomassie stained gel and an anti-His-tag immunoblot of Tp4 

44 expression, using a bacterial expression vector; and, FIG. 21(b) is a photograph of a 

45 Coomassie stained gel and an anti-His-tag immunoblot of Tp5 expression, using a 

46 bacterial expression vector. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



4 



(41860-189496) 

1 

2 

3 FIG. 22 is a panel of histograms indicating numbers of infected Bovine lymphocytes, 

4 stained with the reporter molecules, -mouse polyclonal antibody to Tp1 (FIG. 22(b)), 

5 mouse monoclonal antibody to Tp1 (FIG. 22(c)), mouse polyclonal antibody to Tp4 

6 (FIG.22(d)), and mouse polyclonal antibody to Tp5, (FIG. 22(e)). 
7 

8 FIG. 23. shows Tp1 multiple sequence alignments, Amino acid sequence comparison of 

9 a portion (containing the HD6 CTL epitope) of Tp1 generated from T. parva isolates from 

10 different regions. Domains with variations are underlined. 

11 
12 

13 DETAILED DESCRIPTION OF THE INVENTION 

14 

15 Class I MHC-restricted CD8 + cytotoxic T lymphocytes (CTLs) are responsible for 

16 protecting cattle against a lethal challenge with 7. parva sporozoites (several reviews 

17 cited). These CTLs are directed at schizont-infected cells, which they recognize- and lyse. 

18 Schizont antigens that are recognized by these CD8 + CTLs are the prime candidates for 

19 inclusion into an effective sub-unit vaccine for ECF. 
20 

21 The present invention provides polynucleotides, and methods for their identification, 

22 which encode useful proteins. The useful proteins encoded by the polynucleotide 

23 sequences of the subject invention are targets of antigen-specific cytotoxic CD8+ T 

24 lymphocytes which have been shown to be protective in adoptive cell transfer 

25 experiments. 
26 

27 
28 
29 
30 
31 

32 In a particularly preferred embodiment of the subject invention, the antigens encoded by 

33 polynucleotide sequences, cloned from expressed genes of Theileria parva, are identified 

34 by the use of immortalized cloned bovine skin fibroblast cell lines. In another preferred 

35 embodiment, stimulation of cytotoxic CD8+ T lymphocytes, by antigen, is measured by 

36 the release of soluble factors, more specifically gamma interferon. The subject invention 

37 also relates to compositions isolated by methods including one or more of these particular 

38 steps and the use of these compositions: to stimulate or induce cytotoxic T cells, as 

39 diagnostic reagents for the detection of disease, or an immune response, in kits or high 

40 throughput "chip" methods for the detection of or expression of, identical or homologous 

41 nucleic acids. 
42 

43 As a result of the degeneracy of the genetic code, a multitude of nucleotide sequences 

44 may be produced which are based upon the sequences provided herein and 

45 corresponding peptides, polypeptides, or proteins. Some of these nucleotide sequences 

46 will bear only minimal homology to the sequences disclosed herein; however the subject 

47 invention specifically contemplates each and every possible variation of nucleotide 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



5 



i 



(41860-189496) 



1 sequence that could be made by selecting combinations based on possible codon 

2 choices. These combinations are made in accordance with the standard triplet genetic 

3 code as applied to the nucleotide sequence of naturally occurring peptide, polypeptide, or 

4 protein, and all such variations are to be considered as being specifically disclosed 

5 herein. 
6 

7 It is possible to produce the polynucleotides of the subject invention, or portions thereof, 

8 entirely by synthetic chemistry. After synthesis, the nucleic acid sequence can be used 

9 alone or joined with a preexisting sequence and inserted into one of the many available 

10 DNA vectors and their respective host cells using techniques well known in the art. 

11 Moreover, synthetic chemistry may be used to introduce specific mutations into the 

12 nucleotide sequence. Alternatively, a portion of sequence in which a mutation is desired 

13 can be synthesized and recombined with a portion of an existing genomic or recombinant 

14 sequence. 
15 

16 Nucleotide sequences encoding a peptide, polypeptide, or protein may be joined to a 

17 variety of other nucleotide sequences by means of well established recombinant DNA 

18 techniques (Sambrook J. et al. (1989) Molecular Cloning: A Laboratory Manual, Cold 

19 Spring Harbor Laboratory, Cold Spring Harbor, N.Y.; or Ausubel F. M. et al. (1989) 

20 Current Protocols in Molecular Biology, John Wiley & Sons, New York City). Useful 

21 sequences include an assortment of cloning vectors such as plasmids, cosmids, lambda 

22 phage derivatives, phagemids, and the like. Vectors of interest include vectors for 

23 replication, expression, probe generation, sequencing, and the like. In general, vectors of 

24 interest may contain an origin of replication functional in at least one organism, 

25 convenient restriction endonuclease sensitive sites, and selectable markers for one or 

26 more host cell systems. 
27 

28 Another aspect of the subject invention is to provide for hybridization probes which are 

29 capable of hybridizing with naturally occurring antigen sequences or nucleotide 

30 sequences encoding the disclosed peptide, polypeptide, or protein. The stringency of the 

31 hybridization conditions will determine whether the probe identifies only the native 

32 nucleotide sequence or sequences of closely related molecules. If degenerate nucleotide 

33 sequences of the subject invention are used for the detection of related sequences, they 

34 should preferably contain at least 50% of the nucleotides of the sequences presented 

35 herein. 
36 

37 Hybridization probes of the subject invention may be derived from the nucleotide 

38 sequences of the attached List Sequences and the Sequences provided in FIGS. 16-20, 

39 or from surrounding or included genomic sequences comprising untranslated regions 

40 such as promoters, enhancers and introns. Such hybridization probes may be labeled 

41 with appropriate reporter molecules. Means for producing specific hybridization probes 

42 include oligolabelling, nick translation, end-labeling or PCR amplification using a labeled 

43 nucleotide. Alternatively, the cDNA sequence may be cloned into a vector for the 

44 production of mRNA probe. Such vectors are known in the art, are commercially 

45 available, and may be used to synthesize RNA probes in vitro by addition of an 

46 appropriate RNA polymerase such as T7, T3 or SP6 and labelled nucleotides. A number 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



6 



(41860-189496) 



1 of companies (such as Pharmacia Biotech, Piscataway, N.J.; Promega, Madison, Wis.; 

2 US Biochemical Corp, Cleveland, Ohio; etc.) supply commercial kits and protocols for 

3 these procedures. 

4 

5 The nucleotide sequences (shown in FIGS. 16-20,) can be used to generate probes for 

6 mapping the native genomic sequence. The sequence may be mapped to a particular 

7 chromosome or to a specific region of the chromosome using well known techniques. 

8 These include in situ hybridization to chromosomal spreads, flow-sorted chromosomal 

9 preparations, or artificial chromosome constructions such as yeast artificial chromosomes 

10 (YACs), bacterial artificial chromosomes (BACs), bacterial P1 constructions or single 

1 1 chromosome cDNA libraries. 
12 

13 In situ hybridization of chromosomal preparations and physical mapping techniques such 

14 as linkage analysis using established chromosomal markers are invaluable in extending 

15 genetic maps. The nucleotide sequences of the subject invention may also be used to 

16 detect differences in the chromosomal location of nucleotide sequences due to 

17 translocation, inversion, or recombination. 
18 

19 Other aspects of the invention include use of the disclosed sequences or recombinant 

20 nucleic acids derived therefrom to produce purified peptides. The nucleotide sequences 

21 as disclosed herein may be used to produce an amino acid sequence using well known 

22 methods of recombinant DNA technology. Goeddel (Gene Expression Technology, 

23 Methods and Enzymology [1990] Vol 185, Academic Press, San Diego, Calif.) is one 

24 among many publications which teach expression of an isolated, purified nucleotide 

25 sequence. The amino acid or peptide may be expressed in a variety of host cells, either 

26 prokaryotic or eukaryotic. Host cells may be from the same species from which the 

27 nucleotide sequence was derived or from a different species. 
28 

29 Still further aspects of the invention use these purified peptides to produce antibodies or 

30 other molecules able to bind to the peptides. These antibodies or binding agents can then 

31 be used for the screening of cells in order to localize the cellular distribution of the 

32 peptides or proteins. The antibodies are also useful for the affinity purification of 

33 recombinantly produced peptides or proteins. Such antibodies are also useful as 

34 diagnostic reagents for the detection of protozoan diseases or in antibody-mediated tests 

35 and assays. 
36 

37 The disclosed nucleotide sequences can be used individually, or in panels, in tests or 

38 assays to detect levels of peptide, polypeptide, or protein expression. The form of such 

39 qualitative or quantitative methods may include northern analysis, dot blot or other 

40 membrane based technologies, dip stick, pin or chip technologies, PCR, ELISAs or other 

41 multiple sample format technologies. 
42 

43 As used herein, the following definitions apply: 
44 

45 An "oligonucleotide" or "oligomer" is a stretch of nucleotide residues which has a 

46 sufficient number of bases to be used in a polymerase chain reaction (PCR). These short 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



7 



(41860-189496) 



1 sequences are based on (or designed from) genomic or cDNA sequences and are used 

2 to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA 

3 or RNA in a particular cell or tissue. Oligonucleotides or oligomers comprise portions of a 

4 DNA sequence having at least about 1 0 nucleotides and as many as about 50 

5 nucleotides, preferably about 15 to 30 nucleotides. They can be chemically synthesized 

6 and may be used as probes. 
7 

8 By the term "recombinant nucleic acid" herein is meant nucleic acid, originally formed in 

9 vitro or in a cell in culture, in general, by the manipulation of nucleic acid by 

10 endonucleases and/or exonucleases and/or polymerases and/or ligases and/or 

11 recombinases, to produce a nucleic acid not normally found in nature. Thus an isolated 

12 nucleic acid, in a linear form, or an expression vector formed in vitro by ligating DNA 

13 molecules that are not normally joined, are both considered recombinant for the purposes 

14 of this invention. It is understood that once a recombinant nucleic acid is made and 

15 reintroduced into a host cell or organism, it will replicate non-recombinantly, i.e., using the 

16 in vivo cellular machinery of the host cell rather than in vitro manipulations; however, such 

17 nucleic acids, once produced recombinantly, although subsequently replicated non- 
18 recombinantly, are still considered recombinant for the purposes of the invention. 
19 

20 "Probes" are nucleic acid sequences of variable length, preferably between at least about 

21 10 and as many as about 6,000 nucleotides, depending on use. They are used in the 

22 detection of identical, similar, or complementary nucleic acid sequences. Longer length 

23 probes are usually obtained from a natural or recombinant source, are highly specific and 

24 much slower to hybridize than oligomers. They may be single- or double-stranded and 

25 designed to have specificity in PCR, hybridization membrane-based, or ELISA-like 

26 technologies. 
27 

28 "Reporter" molecules are chemical moieties used for labeling a nucleic or amino acid 

29 sequence. They include, but are not limited to, radionuclides, enzymes, fluorescent, 

30 chemi-luminescent, or chromogenic agents. Reporter molecules associate with, establish 

31 the presence of, and may allow quantification of a particular nucleic or amino acid 

32 sequence. 
33 

34 A "portion" or "fragment" of a polynucleotide or nucleic acid comprises all or any part of 

35 the nucleotide sequence having fewer nucleotides than about 6 kb, preferably fewer than 

36 about 1 kb which can be used as a probe. Such probes may be labeled with reporter 

37 molecules using nick translation, Klenow fill-in reaction, PCR or other methods well 

38 known in the art. After pretesting to optimize reaction conditions and to eliminate false 

39 positives, nucleic acid probes may be used in Southern, northern or in situ hybridizations 

40 to determine whether target DNA or RNA is present in a biological sample, cell type, 

41 tissue, organ or organism. 
42 

43 A "polypeptide" comprises a protein, oligopeptide or peptide fragments thereof. 
44 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



8 



(41860-189496) 



1 A mutant, variant, or modified polypeptide means any polypeptide encoded by a 

2 nucleotide sequence that has been mutated through insertions, deletions, substitutions, or 

3 the like. 
4 

5 "Recombinant nucleotide variants" are alternate polynucleotides which encode a 

6 particular protein. They may be synthesized, for example, by making use of the 

7 "redundancy" in the genetic code. Various codon substitutions, such as the silent changes 

8 which produce specific restriction sites or codon usage-specific mutations, may be 

9 introduced to optimize cloning into a plasmid or viral vector or expression in a particular 
10 prokaryotic or eukaryotic host system, respectively. 

11 

12 "Linkers" are synthesized palindromic nucleotide sequences which create internal 

13 restriction endonuclease sites for ease of cloning the genetic material of choice into 

14 various vectors. "Polylinkers" are engineered to include multiple restriction enzyme sites 

15 and provide for the use of both those enzymes which leave 5' and 3' overhangs such as 

16 BamHI, EcoRI, Pstl, Kpnl and Hind III or which provide a blunt end such as EcoRV, 

17 SnaBI and Stul. 
18 

19 

20 "Chimeric" molecules are polynucleotides or polypeptides which are created by combining 

21 one or more nucleotide peptide sequences (or their parts). In the case of nucleotide 

22 sequences, such combined sequences may be introduced into an appropriate vector and 

23 expressed to give rise to a chimeric polypeptide which may be expected to be different 

24 from the native molecule in one or more of the following characteristics: cellular location, 

25 distribution, ligand-binding affinities, interchain affinities, degradation/turnover rate, 

26 signaling, etc. 
27 

28 "Active" is that state which is capable of being useful or of carrying out some role. It 

29 specifically refers to those forms, fragments, or domains of an amino acid sequence 

30 which display the biologic and/or immunogenic activity characteristic of the naturally 

31 occurring peptide, polypeptide, or protein. 

32 

33 "Naturally occurring" refers to a polypeptide produced by cells which have not been 

34 genetically engineered or which have been genetically engineered to produce the same 

35 sequence as that naturally produced. 

36 

37 "Derivative" refers to those polypeptides which have been chemically modified by such 

38 techniques as ubiquitination, labeling, pegylation (derivatization with polyethylene glycol), 

39 and chemical insertion or substitution of amino acids such as ornithine which do not 

40 normally occur in proteins. 
41 

42 "Recombinant polypeptide variant" refers to any polypeptide which differs from naturally 

43 occurring peptide, polypeptide, or protein by amino acid insertions, deletions and/or 

44 substitutions. 
45 

46 Amino acid "substitutions" are defined as one for one amino acid replacements. They are 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



9 



(41860-189496) 

1 conservative in nature when the substituted amino acid has similar structural and/or 

2 chemical properties. Examples of conservative replacements are substitution of a leucine 

3 with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine. 
4 

5 Amino acid "insertions" or "deletions" are changes to or within an amino acid sequence. 

6 They typically fall in the range of about 1 to 5 amino acids. The variation allowed in a 

7 particular amino acid sequence may be experimentally determined by producing the 

8 peptide synthetically or by systematically making insertions, deletions, or substitutions of 

9 nucleotides in the sequence using recombinant DNA techniques. 
10 

11 

12 An "oligopeptide" is a short stretch of amino acid residues and may be expressed from an 

13 oligonucleotide. Such sequences comprise a stretch of amino acid residues of at least 

14 about 5 amino acids and often about 1 7 or more amino acids, typically at least about 9 to 

15 13 amino acids, and of sufficient length to display biologic and/or immunogenic activity. 
16 

17 

18 [0061] A "standard" is a quantitative or qualitative measurement for comparison. 

19 Preferably, it is based on a statistically appropriate number of samples and is created to 

20 use as a basis of comparison when performing diagnostic assays, running clinical trials, 

21 or following patient treatment profiles. The samples of a particular standard may be 

22 normal or similarly abnormal. 
23 

24 An antibody or "specific binding parts" means any fragment of an antibody molecule that 

25 will bind antigen or other ligands such as lectins or other molecules. "Specific binding 

26 parts" is meant to include, but not be limited to antibody fragments such as Fab 

27 fragments, Fab'(2) fragments, Fc region fragments, Complimentarity determing regions 

28 (CDRs), Fv fragments, single chain Fv (scFv) fragments, and antigen binding site 

29 fragments. 

30 J 

31 AT cell "epitope" is an antigenic determinant recognized and bound by the T-cell 

32 receptor. Epitopes recognized by the T-cell receptor are often located in the inner, 

33 unexposed side of the antigen, and become accessible to the T-cell receptors after 

34 proteolytic processing of the antigen. 

35 

36 An "ELISpot Assay: Short for Enzyme-linked ImmunoSpot Assay", means an antibody 

37 based method to detect secretion of soluble factors released by cells. Originally 

38 developed as a method to detect antibody-secreting B-cells, later the method was 

39 adapted to determine T-cell reaction to a specific antigen, often represented as number 

40 of activated cells per million. 
41 

42 A Bovine MHC class II bioassay, means an assay measures IFN-y release from T cells 

43 responding to specific stimulation through the ability of IFN-y to induce and up-regulate 

44 expression of class II molecules on bovine endothelial cells. Bovine endothelial cells do 

45 not constutively express class II molecules unless triggered by external signals such as 

46 IFN-y. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



10 



(41860-189496) 



1 

2 Since the list of technical and scientific terms cannot be all encompassing, any undefined 

3 terms shall be construed to have the same meaning as is commonly understood by one 

4 of skill in the art to which this invention belongs. Furthermore, the singular forms "a", "an" 

5 and "the" include plural referents unless the context clearly dictates otherwise. 

6 

7 The invention is not to be limited only to the particular sequences, variants, formulations 

8 or methods described. The sequences, variants, formulations and methodologies may 

9 vary, and the terminology used herein is for the purpose of describing particular 
10 embodiments. The terminology and definitions are not intended to be limiting. 
11 

12 Polynucleotide probes employing the present inventive compositions. DNA possesses a 

13 fundamental property called base complementarity. In nature, DNA ordinarily exists in the 

14 form of pairs of anti-parallel strands, the bases on each strand projecting from that strand 

15 toward the opposite strand. The base adenine (A) on one stand will always be opposed 

16 to the base thymine (T) on the other strand, and the base guanine (G) will be opposed to 

17 the base cytosine (C). The bases are held it apposition by their ability to hydrogen bond 

18 in this specific way. Though each individual bond is relatively weak, the net effect of 

19 many adjacent hydrogen bonded bases, together with base stacking effects, is a stable 

20 joining of the two complementary strands. These bonds can be broken by treatments 

21 such as high pH or high temperature, and these conditions result in the dissociation, or 

22 "denaturation," of the two strands. If the DNA is then placed in conditions which make 

23 hydrogen bonding of the bases thermodynamically favorable, the DNA strands will 

24 anneal, or "hybridize," and reform the original double-stranded DNA. If carried out under 

25 appropriate conditions, this hybridization can be highly specific. That is, only strands with 

26 a high degree of base complementarity will be able to form stable double-stranded 

27 structures. The relationship of the specificity of hybridization to reaction conditions is well 

28 known. Thus, hybridization may be used to test whether two pieces of DNA are 

29 complementary in their base sequences. It is this hybridization mechanism which 

30 facilitates the use of probes of the subject invention to readily detect and characterize 

3 1 DNA sequences of interest. 
32 

33 The specifically exemplified polynucleotides of the subject invention can themselves be 

34 used as probes. Additional polynucleotide sequences can be added to the ends of (or 

35 internally in) the exemplified polynucleotide sequences so that polynucleotides that are 

36 longer than the exemplified polynucleotides can also be used as probes. Thus, isolated 

37 polynucleotides comprising one or more of the exemplified sequences are within the 

38 scope of the subject invention. Polynucleotides that have less nucleotides than the 

39 exemplified polynucleotides can also be used and are contemplated within the scope of 

40 the present invention. For example, for some purposes, it might be useful to use a 

41 conserved sequence from an exemplified polynucleotide wherein the conserved 

42 sequence comprises a portion of an exemplified sequence. Thus, polynucleotides of the 

43 subject invention can be used to find additional, homologous (wholly or partially) genes. 
44 

45 Probes of the subject invention may be composed of DNA, RNA, or PNA (peptide nucleic 

46 acid). The probe will normally have at least about 10 bases, more usually at least about 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



11 



(41860-189496) 



1 17 bases, and may have up to about 100 bases or more. Longer probes can readily be 

2 utilitzed, and such probes can be, for example, several kilobases in length. The probe 

3 sequence is designed to be at least substantially complementary to a portion of a gene 

4 encoding a protein of interest. The probe need not have perfect complementarity to the 

5 sequence to which it hybridizes. The probes may be labeled utilizing techniques that are 

6 well known to those skilled in this art. 
7 

8 One approach for the use of the subject invention as probes entails first identifying DNA 

9 segments that are homologous with the disclosed nucleotide sequences using, for 

10 example, Southern blot analysis of a gene bank. Thus, it is possible, without the aid of 

1 1 biological analysis, to know in advance the probable activity of many new 

12 polynucleotides, and of the individual gene products expressed by a given 

13 polynucleotide. Such an analysis provides a rapid method for identifying commercially 

14 valuable compositions. 
15 

16 One hybridization procedure useful according to the subject invention typically includes 

17 the initial steps of isolating the DNA sample of interest and purifying it chemically. Either 

18 lysed cells or total fractionated nucleic acid isolated from cells can be used. Cells can be 

19 treated using known techniques to liberate their DNA (and/or RNA). The DNA sample 

20 can be cut into pieces with an appropriate restriction enzyme. The pieces can be interest 

21 can be through electrophoresis in a gel, usually agarose or acrylamide. The pieces of 

22 interest can be transferred to an immobilizing membrane. 
23 

24 The particular hybridization technique is not essential to the subject invention. As 

25 improvements are made in hybridization techniques, they can be readily applied. 

26 

27 The probe and sample can then be combined in a hybridization buffer solution and held 

28 at an appropriate temperature until annealing occurs. Thereafter, the membrane is 

29 washed free of extraneous materials, leaving the sample and bound probe molecules 

30 typically detected and quantified by autoradiography and/or liquid scintillation counting. 

31 As is well known in the art, if the probe molecule and nucleic acid sample hybridize by 

32 forming a strong non-covalent bond between the two molecules, it can be reasonably 

33 assumed that the probe and sample are essentially identical or very similar. The probe's 

34 detectable label provides a means for determining in a known manner whether 

35 hybridization has occurred. 
36 

37 In the use of the nucleotide segments as probes, the particular probe is labeled with any 

38 suitable label known to those skilled in the art, including radioactive and non-radioactive 

39 labels. Typical radioactive labels include .sup.32P, 35S, or the like. Non-radioactive 

40 labels include, for example, ligands such as biotin or thyroxine, as well as enzymes such 

41 as hydrolases or peroxidases, or the various chemiluminescers such as luciferin, or 

42 fluorescent compounds like fluorescein and its derivatives. In addition, the probes can be 

43 made inherently fluorescent as described in International Application No. WO 93/16094. 
44 

45 Various degrees of stringency of hybridization can be employed. The more stringent the 

46 conditions, the greater the complementarity that is required for duplex formation. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



12 



I 



(41860-189496) 



1 Stringency can be controlled by temperature, probe concentration, probe length, ionic 

2 strength, time, and the like. Preferably, hybridization is conducted under moderate to 

3 high stringency conditions by techniques well known in the art, as described, for 

4 example, in Keller, G. H., M. M. Manak (1987) DNA Probes, Stockton Press, New York, 

5 N.Y., pp. 169-170. 
6 

7 As used herein "moderate to high stringency" conditions for hybridization refers to 

8 conditions that achieve the same, or about the same, degree of specificity of 

9 hybridization as the conditions "as described herein." Examples of moderate to high 

10 stringency conditions are provided herein. Specifically, hybridization of immobilized DNA 

1 1 on Southern blots with .sup.32P-labeled gene-specific probes was performed using 

12 standard methods (Maniatis et al.). In general, hybridization and subsequent washes 

13 were carried out under moderate to high stringency conditions that allowed for detection 

14 of target sequences with homology to sequences exemplified herein. For double- 

15 stranded DNA gene probes, hybridization was carried out overnight at 20-25.degree. C. 

16 below the melting temperature (Tm) of the DNA hybrid in 6.times. SSPE, S.times. 

17 Denhardt's solution, 0.1% SDS, 0.1 mg/ml denatured DNA. The melting temperature is 

18 described by the following formula from Beltz et al. (1983): 
19 

20 Tm=81.5.degree. C.+16.6 Log[Na+]+0.41(% G+C)-0.61(% formamide)-600/length of 

21 duplex in base pairs. 

22 

23 Washes are typically carried out as follows: 
24 

25 (1 ) Twice at room temperature for 1 5 minutes in 1 .times. SSPE, 0.1 % SDS (low 

26 stringency wash). 

27 

28 (2) Once at Tm-20.degree, C. for 15 minutes in 0.2.times. SSPE, 0.1% SDS (moderate 

29 stringency wash). 

30 

31 For oligonucleotide probes, hybridization was carried out overnight at 10-20.degree. C. 

32 below the melting temperature (Tm) of the hybrid in 6.times. SSPE, S.times. Denhardt's 

33 solution, 0.1% SDS, 0.1 mg/ml denatured DNA. Tm for oligonucleotide probes was 

34 determined by the following formula from Suggs et al. (1981 ): 

35 

36 Tm (.degree. C.)=2(number T/A base pairs)+4(number G/C base pairs) 

37 

38 Washes were typically carried out as follows: 
39 

40 (1 ) Twice at room temperature for 1 5 minutes 1 .times. SSPE, 0.1 % SDS (low stringency 

41 wash). 
42 

43 (2) Once at the hybridization temperature for 15 minutes in 1 .times. SSPE, 0.1% SDS 

44 (moderate stringency wash). 
45 

46 In general, salt and/or temperature can be altered to change stringency. With a labeled 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



13 



I 



(41860-189496) 



1 DNA fragment of greater than about 70 or so bases in length, the following conditions 

2 can be used: 

3 

4 1 Low: 1 or 2X SSPE, room temperature Low: 1 or 2X SSPE, 42.degree. C. Moderate: 

5 0.2X or 1X SSPE, 65.degree. C. High: 0.1X SSPE, 65.degree. C. 

6 

7 Duplex formation and stability depend on substantial complementarity between the two 

8 strands of a hybrid, and, as noted above, a certain degree of mismatch can be tolerated. 

9 Therefore, polynucleotide sequences of the subject invention include mutations (both 

10 single and multiple), deletions, and insertions in the described sequences, and 

1 1 combinations thereof, wherein said mutations, insertions, and deletions permit formation 

12 of stable hybrids with a target polynucleotide of interest. Mutations, insertions, and 

13 deletions can be produced in a given polynucleotide sequence using standard methods 

14 known in the art. Other methods may become known in the future. 
15 

16 The mutational, insertional, and deletional variants of the polypeptide sequences of the 

17 invention can be used in the same manner as the exemplified polynucleotide sequences 

18 so long as the variants have substantial sequence similarity with the original sequence. 

19 As used herein, substantial sequence similarity refers to the extent of nucleotide 

20 similarity that is sufficient to enable the variant polynucleotide to function in the same 

21 capacity as the original sequence. Preferably, this similarity is greater than 50%; more 

22 preferably, this similarity is greater than 75%; and most preferably, this similarity is 

23 greater than 90%. The degree of similarity needed for the variant to function in its 

24 intended capacity will depend upon the intended use of the sequence. It is well within the 

25 skill of a person trained in this art to make mutational, insertional, and deletional 

26 mutations that are designed to improve the function of the sequence or otherwise provide 

27 a methodological advantage. 
28 

29 PCR technology. Polymerase Chain Reaction (PGR) is a repetitive, enzymatic, primed 

30 synthesis of a nucleic acid sequence. This procedure is well known and commonly used 

31 by those skilled in this art (see U.S. Pat. Nos. 4,683,195, 4,683,202, and 4,800,159; Saiki 

32 et al., 1985). PCR is based on the enzymatic amplification of a DNA fragment of interest 

33 that is flanked by two oligonucleotide primers that hybridize to opposite strands of the 

34 target sequence. The primers are oriented with the 3' ends pointing towards each other. 

35 Repeated cycles of heat denaturation of the template, annealing of the primers to their 

36 complementary sequences, and extension of the annealed primers with a DNA 

37 polymerase result in the amplification of the segment defined by the 5' ends of the PCR 

38 primers. Since the extension product of each primer can serve as a template for the other 

39 primer, each cycle essentially doubles the amount of DNA fragment produced in the 

40 previous cycle. This results in the exponential accumulation of the specific target 

41 fragment, up to several million-fold in a few hours. By using a thermostable DNA 

42 polymerase such as Taq polymerase, which is isolated from the thermophilic bacterium 

43 Thermus aquaticus, the amplification process can be completely automated. Other 

44 enzymes that can be used are known to those skilled in the art. 
45 

46 The polynucleotide sequences of the subject invention (and portions thereof such as 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



14 



(41860-189496) 



1 conserved regions and portions that serve to distinguish these sequences from 

2 previously-known sequences) can be used as, and/or used in the design of, primers for 

3 PCR amplification. In performing PCR amplification, a certain degree of mismatch can be 

4 tolerated between primer and template. Therefore, mutations, deletions, and insertions 

5 (especially additions of nucleotides to the 5' end) of the exemplified polynucleotides can 

6 be used in this manner. Mutations, insertions and deletions can be produced in a given 

7 primer by methods known to an ordinarily skilled artisan. 
8 

9 Full length genes may be cloned utilizing partial nucleotide sequence and various 

10 methods known in the art. Gobinda et al. (1993; PCR Methods Applic 2:318-22) disclose 

1 1 "restriction-site PCR" as a direct method which uses universal primers to retrieve 

12 unknown sequence adjacent to a known locus. First, genomic DNA is amplified in the 

13 presence of primer to linker and a primer specific to the known region. The amplified 

14 sequences are subjected to a second round of PCR with the same linker primer and 

15 another specific primer internal to the first one. Products of each round of PCR are 

16 transcribed with an appropriate RNA polymerase and sequenced using reverse 

17 transcriptase. 
18 

19 Inverse PCR can be used to acquire unknown sequences starting with primers based on 

20 a known region (Triglia T. et al. (1988) Nucleic Acids Res 16:8186). The method uses 

21 several restriction enzymes to generate a suitable fragment in the known region of a 

22 gene. The fragment is then circularized by intramolecular ligation and used as a PCR 

23 template. Divergent primers are designed from the known region. The multiple rounds of 

24 restriction enzyme digestions and ligations that are necessary prior to PCR make the 

25 procedure slow and expensive (Gobinda et al. [1993] supra). 
26 

27 Capture PCR (Lagerstrom M. etal. (1991) PCR Methods Applic 1:111-19) is a method 

28 for PCR amplification of DNA fragments adjacent to a known sequence in eucaryotic and 

29 YAC DNA. As noted by Gobinda et al. (1993,supra), capture PCR also requires multiple 

30 restriction enzyme digestions and ligations to place an engineered double-stranded 

31 sequence into an unknown portion of the DNA molecule before PCR. Although the 

32 restriction and ligation reactions are carried out simultaneously, the requirements for 

33 extension, immobilization and two rounds of PCR and purification prior to sequencing 

34 render the method cumbersome and time consuming. 
35 

36 Parker J. D. et al. (Nucleic Acids Res [1991 1 19:3055-60), teach walking PCR, a method 

37 for targeted gene walking which permits retrieval of unknown sequences. 

38 PromoterFinder.TM. is a kit available from Clontech Laboratories, Inc. (Palo Alto, Calif.) 

39 which uses PCR and primers derived from p53 to walk in genomic DNA. Nested primers 

40 and special PromoterFinder.TM. libraries are used to detect upstream sequences such 

41 as promoters and regulatory elements. This process avoids the need to screen libraries 

42 and is useful in finding intron/exon junctions. 
43 

44 One PCR method replaces methods which use labeled probes to screen plasmid 

45 libraries and allow one researcher to process only about 3-5 genes in 14-40 days. In the 

46 first step, which can be performed in about two days, any two of a plurality of primers are 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



15 



(41860-189496) 



1 designed and synthesized based on a known partial sequence. In step 2, which takes 

2 about six to eight hours, the sequence is extended by PCR amplification of a selected 

3 library. Steps 3 and 4, which take about one day, are purification of the amplified cDNA 

4 and its ligation into an appropriate vector. Step 5, which takes about one day, involves 

5 transforming and growing up host bacteria. In step 6, which takes approximately five 

6 hours, PCR is used to screen bacterial clones for extended sequence. The final steps, 

7 which take about one day, involve the preparation and sequencing of selected clones. 
8 

9 If the full length cDNA has not been obtained, the entire procedure is repeated using 

10 either the original library or some other preferred library. The preferred library may be 

1 1 one that has been size-selected to include only larger cDNAs or may consist of single or 

12 combined commercially available libraries, e.g., from Clontech Laboratories, Inc. (Palo 

13 Alto, Calif.). The cDNA library may have been prepared with oligo (dT) or random 

14 priming. Random primed libraries are preferred in that they will contain more sequences 

15 which contain 5* ends of genes. A randomly primed library may be particularly useful if an 

16 oligo (dT) library does not yield a complete gene. It must be noted that the larger and 

17 more complex the protein, the less likely it is that the complete gene will be found in a 

18 single plasmid. 
19 

20 CLONTECH PCR-Select.TM. cDNA Subtraction (Clontech Laboratories, Inc., Palo Alto, 

21 Calif.) is yet another means by which differentially expressed genes may be isolated. The 

22 procedure allows for the isolation of transcripts present in one mRNA population which is 

23 absent, or found in reduced numbers, in a second population of mRNA. Rare transcripts 

24 may be enriched 1000-fold. 
25 

26 Another method for analyzing either the size or the nucleotide sequence of PCR products 

27 is capillary electrophoresis. Systems for rapid sequencing are available from Perkin 

28 Elmer (Foster City Calif.), Beckman Instruments (Fullerton, Calif.), and other companies. 

29 Capillary sequencing employs flowable polymers for electrophoretic separation, four 

30 different fluorescent dyes (one for each nucleotide) which are laser activated, and 

31 detection of the emitted wavelengths by a charge coupled devise camera. Output/light 

32 intensity is converted to electrical signal using appropriate software (eg. GenotyperTM. 

33 and Sequence NavigatorsTM. from Perkin Elmer) and the entire process from loading of 

34 samples to computer analysis and electronic data display is computer controlled. 

35 Capillary electrophoresis provides greater resolution and is many times faster than 

36 standard gel based procedures. It is particularly suited to the sequencing of small pieces 

37 of DNA which might be present in limited amounts in a particular sample. The 

38 reproducible sequencing of up to 350 bp of M13 phage DNA in 30 min has been reported 

39 (Ruiz-Martinez M. C. et al. [1993] Anal Chem 65:2851-8). 
40 

41 Polynucleotides and proteins. Polynucleotides of the subject invention can be defined 

42 according to several parameters. One characteristic is the biological activity of the protein 

43 products as identified herein. The proteins and genes of the subject invention can be 

44 further defined by their amino acid and nucleotide sequences. The sequences of the 

45 molecules can be defined in terms of homology to certain exemplified sequences as well 

46 as in terms of the ability to hybridize with, or be amplified by, certain exemplified probes 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



16 



(41860-189496) 



1 and primers. Additional primers and probes can readily be constructed by those skilled in 

2 the art such that alternate polynucleotide sequences encoding the same amino acid 

3 sequences can be used to identify and/or characterize additional genes. The proteins of 

4 the subject invention can also be identified based on their immunoreactivity with certain 

5 antibodies. 
6 

7 The polynucleotides and proteins of the subject invention include portions, fragments, 

8 variants, and mutants of the full-length sequences as well as fusions and chimerics, so 

9 long as the encoded protein retains the characteristic biological activity of the proteins 

10 identified herein. As used herein, the terms "variants" or "variations" of genes refer to 

1 1 nucleotide sequences that encode the same proteins or which encode equivalent 

12 proteins having equivalent biological activity. As used herein, the term "equivalent 

13 proteins" refers to proteins having the same or essentially the same biological activity as 

14 the exemplified proteins. 
15 

16 Variations of genes may be readily constructed using standard techniques such as site- 

17 directed mutagenesis and other methods of making point mutations and by DNA 

18 shuffling, for example. In addition, gene and protein fragments can be made using 

19 commercially available exonucleases, endonucleases, and proteases according to 

20 standard procedures. For example, enzymes such as Bal31 can be used to 

21 systematically cut off nucleotides from the ends of genes. Also, genes that encode 

22 fragments may be obtained using a variety of restriction enzymes. Proteases may be 

23 used to directly obtain active fragments of these proteins. Of course, molecular 

24 techniques for cloning polynucleotides and producing gene constructs of interest are also 

25 well known in the art. In vitro evaluation techniques, such as MAXYGEN's "Molecular 

26 Breeding" can also be applied to practice the subject invention. 
27 

28 Because of the redundancy of the genetic code, a variety of different DNA sequences 

29 can encode the amino acid sequences encoded by the polynucleotide sequences 

30 disclosed herein. It is well within the skill of a person trained in the art to create these 

31 alternative DNA sequences encoding proteins having the same, or essentially the same, 

32 amino acid sequence. These variant DNA sequences are within the scope of the subject 

33 invention. As used herein, reference to "essentially the same" sequence refers to 

34 sequences that have amino acid substitutions, deletions, additions, or insertions that do 

35 not materially affect biological activity. Fragments retaining the characteristic biological 

36 activity are also included in this definition. 
37 

38 A farther method for identifying genes and polynucleotides (and the proteins encoded 

39 thereby) of the subject invention is through the use of oligonucleotide probes. Probes 

40 provide a rapid method for identifying genes of the subject invention. The nucleotide 

41 segments that are used as probes according to the invention can be synthesized using a 

42 DNA synthesizer and standard procedures. 
43 

44 The subject invention comprises variant or equivalent proteins (and nucleotide 

45 sequences coding for equivalent proteins) having the same or similar biological activity of 

46 proteins encoded by the exemplified polynucleotides. Equivalent proteins will have amino 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



17 



(41860-189496) 



1 acid similarity with an exemplified protein (or peptide). The amino acid identity will 

2 typically be greater than 60%. Preferably, the amino acid identity will be greater than 

3 75%. More preferably, the amino acid identity will be greater than 80%, and even more 

4 preferably greater than 90%. Most preferably, amino acid identity will be greater than 

5 95%. (Likewise, the polynucleotides that encode the subject polypeptides will also have 

6 corresponding identities in these preferred ranges.) These identities are as determined 

7 using standard alignment techniques for determining amino acid identity. The amino acid 

8 identity/similarity/homology will be highest in critical regions of the protein including those 

9 regions that account for biological activity or that are involved in the determination of 

10 three-dimensional configuration that is ultimately responsible for the biological activity. In 

11 this regard, certain amino acid substitutions are acceptable and can be expected if these 

12 substitutions are in regions which are not critical to activity or are conservative amino 

13 acid substitutions which do not affect the three-dimensional configuration of the 

14 molecule. For example, amino acids may be placed in the following classes: non-polar, 

15 uncharged polar, basic, and acidic. Conservative substitutions whereby an amino acid of 

16 one class is replaced with another amino acid of the same type fall within the scope of 

17 the subject invention so long as the substitution does not materially alter the biological 

18 activity of the compound. Below is a list of examples of amino acids belonging to various 

19 classes. 
20 

21 Class of Amino Acid Examples of Amino Acids Nonpolar Ala, Val, Leu, lie, Pro, Met, Phe, 

22 Trp Uncharged Polar Gly, Ser, Thr, Cys, Tyr, Asn, Gin Acidic Asp, Glu Basic Lys, His 
23 

24 In some instances, non-conservative substitutions can also be made. 
25 

26 As used herein, reference to "isolated" polynucleotides and/or "purified" proteins refers to 

27 these molecules when they are not associated with the other molecules with which they 

28 would be found in nature. Thus, reference to "isolated" and/or "purified" signifies the 

29 involvement of the "hand of man" as described herein. Reference to "heterologous" 

30 proteins, genes, and gene constructs, also signifies the involvement of the "hand of 

31 man." 
32 

33 Vectors/Host Cells 
34 

35 The invention also provides vectors containing the nucleic acid molecules described 

36 herein. The term "vector" refers to a vehicle, preferably a nucleic acid molecule, which 

37 can transport the nucleic acid molecules. When the vector is a nucleic acid molecule, the 

38 nucleic acid molecules are covalently linked to the vector nucleic acid. With this aspect of 

39 the invention, the vector includes a plasmid, single or double stranded phage, a single or 

40 double stranded RNA or DNA viral vector, or artificial chromosome, such as a BAC, PAC, 

41 YAC, OR MAC. 
42 

43 A vector can be maintained in the host cell as an extrachromosomal element where it 

44 replicates and produces additional copies of the nucleic acid molecules. Alternatively, the 

45 vector may integrate into the host cell genome and produce additional copies of the 

46 nucleic acid molecules when the host cell replicates. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



18 



(41860-189496) 



1 

2 The invention provides vectors for the maintenance (cloning vectors) or vectors for 

3 expression (expression vectors) of the nucleic acid molecules. The vectors can function 

4 in prokaryotic or eukaryotic cells or in both (shuttle vectors). 
5 

6 Expression vectors contain cis-acting regulatory regions that are operably linked in the 

7 vector to the nucleic acid molecules such that transcription of the nucleic acid molecules 

8 is allowed in a host cell. The nucleic acid molecules can be introduced into the host cell 

9 with a separate nucleic acid molecule capable of affecting transcription. Thus, the second 

10 nucleic acid molecule may provide a trans-acting factor interacting with the cis-regulatory 

1 1 control region to allow transcription of the nucleic acid molecules from the vector. 

12 Alternatively, a trans-acting factor may be supplied by the host cell. Finally, a trans-acting 

13 factor can be produced from the vector itself. It is understood, however, that in some 

14 embodiments, transcription and/or translation of the nucleic acid molecules can occur in 

15 a cell-free system. 
16 

17 The regulatory sequence to which the nucleic acid molecules described herein can be 

18 operably linked include promoters for directing mRNA transcription. These include, but 

19 are not limited to, the left promoter from bacteriophage .lambda., the lac, TRP, and TAC 

20 promoters from E. Coli, the early and late promoters from SV40, the CMV immediate 

21 early promoter, the adenovirus early and late promoters, and retrovirus long-terminal 

22 repeats. 
23 

24 In addition to control regions that promote transcription, expression vectors may also 

25 include regions that modulate transcription, such as repressor binding sites and 

26 enhancers. Examples include the SV40 enhancer, the cytomegalovirus immediate early 

27 enhancer, polyoma enhancer, adenovirus enhancers, and retrovirus LTR enhancers. 

28 

29 In addition to containing sites for transcription initiation and control, expression vectors 

30 can also contain sequences necessary for transcription termination and, in the 

31 transcribed region a ribosome binding site for translation. Other regulatory control 

32 elements for expression include initiation and termination codons as well as 

33 polyadenylation signals. The person of ordinary skill in the art would be aware of the 

34 numerous regulatory sequences that are useful in expression vectors. Such regulatory 

35 sequences are described, for example, in Sambrook et al., Molecular Cloning: A 

36 Laboratory Manual. 2nd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 

37 N.Y., (1989). 
38 

39 A variety of expression vectors can be used to express a nucleic acid molecule. Such 

40 vectors include chromosomal, episomal, and virus-derived vectors, for example vectors 

41 derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast 

42 chromosomal elements, including yeast artificial chromosomes, from viruses such as 

43 baculoviruses, papovaviruses such as SV40, Vaccinia viruses, adenoviruses, poxviruses, 

44 pseudorabies viruses, and retroviruses. Vectors may also be derived from combinations 

45 of these sources such as those derived from plasmid and bacteriophage genetic 

46 elements, e.g. cosmids and phagemids. Appropriate cloning and expression vectors for 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



19 



(41860-189496) 



1 prokaryotic and eukaryotic hosts are described in Sambrook et al., Molecular Cloning: A 

2 Laboratory Manual. 2nd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 

3 N.Y.,(1989). 
4 

5 The regulatory sequence may provide constitutive expression in one or more host cells 

6 (i.e. tissue specific) or may provide for inducible expression in one or more cell types 

7 such as by temperature, nutrient additive, or exogenous factor such as a hormone or 

8 other ligand. A variety of vectors providing for constitutive and inducible expression in 

9 prokaryotic and eukaryotic hosts are well known to those of ordinary skill in the art. 
10 

1 1 The nucleic acid molecules can be inserted into the vector nucleic acid by well-known 

12 methodology. Generally, the DNA sequence that will ultimately be expressed is joined to 

13 an expression vector by cleaving the DNA sequence and the expression vector with one 

14 or more restriction enzymes and then ligating the fragments together. Procedures for 

15 restriction enzyme digestion and ligation are well known to those of ordinary skill in the 

16 art. 
17 

18 The vector containing the appropriate nucleic acid molecule can be introduced into an 

19 appropriate host cell for propagation or expression using well-known techniques. 

20 Bacterial cells include, but are not limited to, E. coli, Streptomyces, and Salmonella 

21 typhimurium. Eukaryotic cells include, but are not limited to, yeast, insect cells such as 

22 Drosophila, animal cells such as COS and CHO cells, and plant cells. 
23 

24 As described herein, it may be desirable to express the peptide as a fusion protein. 

25 Accordingly, the invention provides fusion vectors that allow for the production of the 

26 peptides. Fusion vectors can increase the expression of a recombinant protein, increase 

27 the solubility of the recombinant protein, and aid in the purification of the protein by acting 

28 for example as a ligand for affinity purification. A proteolytic cleavage site may be 

29 introduced at the junction of the fusion moiety so that the desired peptide can ultimately 

30 be separated from the fusion moiety. Proteolytic enzymes include, but are not limited to, 

31 factor Xa, thrombin, and enteroenzyme. Typical fusion expression vectors include pGEX 

32 (Smith et al., Gene 67:31-40 (1988)), pMAL (New England Biolabs, Beverly, Mass.) and 

33 pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), 

34 maltose E binding protein, or protein A, respectively, to the target recombinant protein. 

35 Examples of suitable inducible non-fusion E. coli expression vectors include pTrc 

36 (Amann et al, Gene 69:301-315 (1988)) and pET 1 1d (Studier et al, Gene Expression 

37 Technology: Methods in Enzymology 1 85:60-89 (1 990)). 
38 

39 Recombinant protein expression can be maximized in host bacteria by providing a 

40 genetic background wherein the host cell has an impaired capacity to proteolytically 

41 cleave the recombinant protein. (Gottesman, S., Gene Expression Technology: Methods 

42 in Enzymology 185, Academic Press, San Diego, Calif. (1990) 1 19-128). Alternatively, 

43 the sequence of the nucleic acid molecule of interest can be altered to provide 

44 preferential codon usage for a specific host cell, for example E. coli. (Wada et al., Nucleic 

45 Acids Res. 20:2111-2118 (1992)). 
46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



20 



(41860-189496) 



1 The nucleic acid molecules can also be expressed by expression vectors that are 

2 operative in yeast. Examples of vectors for expression in yeast e.g., S. cerevisiae include 

3 pYepSecl (Baldari, et al, EMBO J. 6:229-234 (1987)), pMFa (Kujan et al, Cell 30:933- 

4 943(1982)), pJRY88 (Schultz et al, Gene 54:1 13-123 (1987)), and pYES2 (Invitrogen 

5 Corporation, San Diego, Calf.). 
6 

7 The nucleic acid molecules can also be expressed in insect cells using, for example, 

8 baculovirus expression vectors. Baculovirus vectors available for expression of proteins 

9 in cultured insect cells (e.g., Sf9 cells) include the pAc series (Smith et al, Mol. Cell Biol. 
10 3:2156-2165 (1983)) and the pVL series (Lucklow et al, Virology 170:31-39 (1989)). 

11 

12 In certain embodiments of the invention, the nucleic acid molecules described herein are 

13 expressed in mammalian cells using mammalian expression vectors. Examples of 

14 mammalian expression vectors include pCDM8 (Seed, B. Nature 329:840(1987)) and 

15 pMT2PC (Kaufman et al., EMBO J. 6:187-195 (1987)). 
16 

17 The expression vectors listed herein are provided by way of example only of the well- 

18 known vectors available to those of ordinary skill in the art that would be useful to 

19 express the nucleic acid molecules. The person of ordinary skill in the art would be aware 

20 of other vectors suitable for maintenance propagation or expression of the nucleic acid 

21 molecules described herein. These are found for example in Sambrook, J., Fritsh, E. F., 

22 and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor 

23 Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. 
24 

25 The invention also encompasses vectors in which the nucleic acid sequences described 

26 herein are cloned into the vector in reverse orientation, but operably linked to a 

27 regulatory sequence that permits transcription of antisense RNA. Thus, an antisense 

28 transcript can be produced to all, or to a portion, of the nucleic acid molecule sequences 

29 described herein, including both coding and non-coding regions. Expression of this 

30 antisense RNA is subject to each of the parameters described above in relation to 

31 expression of the sense RNA (regulatory sequences, constitutive or inducible expression, 

32 tissue-specific expression). 
33 

34 The invention also relates to recombinant host cells containing the vectors described 

35 herein. Host cells therefore include prokaryotic cells, lower eukaryotic cells such as 

36 yeast, other eukaryotic cells such as insect cells, and higher eukaryotic cells such as 

37 mammalian cells. 

38 

39 The recombinant host cells are prepared by introducing the vector constructs described 

40 herein into the cells by techniques readily available to the person of ordinary skill in the 

41 art. These include, but are not limited to, calcium phosphate transfection, DEAE-dextran- 

42 mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, 

43 infection, lipofection, and other techniques such as those found in Sambrook, et al. 

44 (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold 

45 Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989). 
46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



21 



(41860-189496) 



1 Host cells can contain more than one vector. Thus, different nucleotide sequences can 

2 be introduced on different vectors of the same cell. Similarly, the nucleic acid molecules 

3 can be introduced either alone or with other nucleic acid molecules that are not related to 

4 the nucleic acid molecules such as those providing trans-acting factors for expression 

5 vectors. When more than one vector is introduced into a cell, the vectors can be 

6 introduced independently, co-introduced or joined to the nucleic acid molecule vector. 
7 

8 In the case of bacteriophage and viral vectors, these can be introduced into cells as 

9 packaged or encapsulated virus by standard procedures for infection and transduction. 

10 Viral vectors can be replication-competent or replication-defective. In the case in which 

11 viral replication is defective, replication will occur in host cells providing functions that 

12 complement the defects. 
13 

14 Vectors generally include selectable markers that enable the selection of the 

15 subpopulation of cells that contain the recombinant vector constructs. The marker can be 

16 contained in the same vector that contains the nucleic acid molecules described herein 

17 or may be on a separate vector. Markers include tetracycline or ampicillin-resistance 

18 genes for prokaryotic host cells and dihydrofolate reductase or neomycin resistance for 

19 eukaryotic host cells. However, any marker that provides selection for a phenotypic trait 

20 will be effective. 
21 

22 While the mature proteins can be produced in bacteria, yeast, mammalian cells, and 

23 other cells under the control of the appropriate regulatory sequences, cell- free 

24 transcription and translation systems can also be used to produce these proteins using 

25 RNA derived from the DNA constructs described herein. 

26 

27 Where secretion of the peptide is desired, which is difficult to achieve with multi- 

28 transmembrane domain containing proteins such as MHC Class l-binding peptides, 

29 appropriate secretion signals are incorporated into the vector. The signal sequence can 

30 be endogenous to the peptides or heterologous to these peptides. 
31 

32 The expressed protein can be isolated from the host cell by standard disruption 

33 procedures, including freeze thaw, sonication, mechanical disruption, use of lysing 

34 agents and the like. The peptide can then be recovered and purified by well-known 

35 purification methods including ammonium sulfate precipitation, acid extraction, anion or 

36 cationic exchange chromatography, phosphocellulose chromatography, hydrophobic- 

37 interaction chromatography, affinity chromatography, hydroxy la patite chromatography, 

38 lectin chromatography, or high performance liquid chromatography. 
39 

40 It is also understood that depending upon the host cell in recombinant production of the 

41 peptides described herein, the peptides can have various glycosylation patterns, 

42 depending upon the cell, or maybe non-glycosylated as when produced in bacteria. In 

43 addition, the peptides may include an initial modified methionine in some cases as a 

44 result of a host-mediated process. 
45 

46 There are many methods for introducing a heterologous gene or polynucleotide into a 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



22 



(41860-189496) 



1 host cell or cells under conditions that allow for stable maintenance and expression of the 

2 gene or polynucleotide. These methods are well known to those skilled in the art. 

3 Synthetic genes, such as, for example, those genes modified to enhance expression in a 

4 heterologous host (such as by preferred codon usage or by the use of adjoining, 

5 downstream, or upstream enhancers) that are functionally equivalent to the genes (and 

6 which encode equivalent proteins) can also be used to transfect hosts. Methods for the 

7 production of synthetic genes are known in the art. Recombinant hosts can be used for 

8 the expression or propagation of genes and polynucleotides of the present invention. The 

9 genes and polynucleotides within the scope of the present invention can be introduced 

10 into a wide variety of microbial or plant hosts, such as bacterial cells, yeast, insect cells, 

1 1 plant cell cultures or plants, mammalian cells. For example, T. parva nucleic acids can 

12 be expressed in a recombinant baculovirus (BV) possessing an optimized promoter and 

13 translation initiation region operably linked to the T. parva nucleic acid. In a preferred 

14 embodiment, an optimized promoter and translation initiation region are operably linked 

15 to the T. parva nucleic acid. In one embodiment, insect host cells can be transformed 

16 with baculovirus expressing T. parva nucleic acids. In still another embodiment, Tn5 

17 (Trichoplusia ni or High Five.TM.) host cells can be transformed with baculovirus 

18 expressing T. parva nucleic acids. 
19 

20 T. parva "recombinant protein", is a protein made using recombinant techniques, i.e. 

21 through the expression of a recombinant nucleic acid as depicted above. A recombinant 

22 protein is distinguished from naturally occurring protein by at least one or more 

23 characteristics. For example, the protein may be isolated or purified away from some or 

24 all of the proteins and compounds with which it is normally associated in its wild type 

25 host, and thus may be substantially pure. For example, an isolated protein is 

26 unaccompanied by at least some of the material with which it is normally associated in its 

27 natural state, preferably constituting at least about 0.5%, more preferably at least about 

28 5% by weight of the total protein in a given sample. A substantially pure protein 

29 comprises at least about 75% by weight of the total protein, with at least about 80% 

30 being preferred, and at least about 90% being particularly preferred. The definition 

31 includes the production of a protein from one organism in a different organism or host 

32 cell. Alternatively, the protein may be made at a significantly higher concentration than is 

33 normally seen, through the use of an inducible promoter or high expression promoter, 

34 such that the protein is made at increased concentration levels. Alternatively, the protein 

35 may be in a form not normally found in nature, as in the addition of an epitope tag or 

36 amino acid substitutions, insertions and/or deletions, as discussed below. 
37 

38 Included in the definition of T. parva antigen polypeptides are T. parva polypeptide 

39 variants. These variants fall into one or more of three classes: substitutional, insertional 

40 or deletional variants. These variants ordinarily are prepared by site specific mutagenesis 

41 of nucleotides in the DNA encoding a T. parva antigen polypeptide, using cassette or 

42 PCR mutagenesis, scanning mutagenesis, gene shuffling or other techniques well known 

43 in the art, to produce DNA encoding the variant, and thereafter expressing the DNA in 

44 recombinant cell culture as outlined above. However, variant T. parva polypeptide 

45 fragments having up to about 100-150 residues may be prepared by in vitro synthesis 

46 using established techniques. Amino acid sequence variants are characterized by the 

47 predetermined nature of the variation, a feature that sets them apart from naturally 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



23 



(41860-189496) 



1 occurring allelic or interspecies variation of the T. parva antigen polypeptide amino acid 

2 sequence. 

3 

4 While the site or region for introducing an amino acid sequence variation is 

5 predetermined, the mutation per se need not be predetermined. For example, in order to 

6 optimize the performance of a mutation at a given site, random mutagenesis may be 

7 conducted at the target codon or region and the expressed T. parva antigen polypeptide 

8 variants can be screened for the optimal combination of desired activity. Techniques for 

9 making mutations at predetermined sites in DNA having a known sequence are well 

10 known. For example, the variations can be made using oligonucleotide-mediated site- 

11 directed mutagenesis [Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. 

12 Acids Res., 10:6487 (1987)], cassette mutagenesis [Wells et al., Gene, 34:315 (1985)], 

13 restriction selection mutagenesis [Wells et al., Philos. Trans. R. Soc. London SerA, 

14 317:415 (1986)], all of which are expressly incorporated by reference, PGR mutagenesis, 

15 or other known techniques can be performed on the cloned DNA to produce T. parva 

16 antigen polypeptide variant DNA. Scanning amino acid analysis can also be employed to 

17 identify one or more amino acids along a contiguous sequence. Among the preferred 

18 scanning amino acids are relatively small, neutral amino acids. Such amino acids include 

19 alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid 

20 among this group because it eliminates the side-chain beyond the beta-carbon and is less 

21 likely to alter the main-chain conformation of the variant [Cunningham and Wells, 

22 Science, 244: 1081-1085 (1989), which is expressly incorporated by reference]. Alanine is 

23 also typically preferred because it is the most common amino acid. Further, it is frequently 

24 found in both buried and exposed positions [Creighton, The Proteins, (W.H. Freeman & 

25 Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976), which are expressly incorporated by 

26 reference]. If alanine substitution does not yield adequate amounts of variant, an isoteric 

27 amino acid can be used. Screening of the mutants or variants is done using Elispot and/or 

28 bioassays of T. parva antigen polypeptide activities and/or properties as described herein. 
29 

30 

31 The present invention further provides fragments of the antigen peptides, in addition to 

32 proteins and peptides that comprise and consist of such fragments, particularly those 

33 comprising the residues identified in FIG. 2. The fragments to which the invention 

34 pertains, however, are not to be construed as encompassing fragments that may be 

35 disclosed publicly prior to the present invention. 
36 

37 As used herein, a fragment comprises at least 8, 10, 12, 14, 16, or more contiguous 

38 amino acid residues from an antigen peptide. Such fragments can be chosen based on 

39 the ability to retain one or more of the biological activities of the antigen peptide or could 

40 be chosen for the ability to perform a function, e.g. bind a substrate or act as an 

41 immunogen. Particularly important fragments are biologically active fragments, peptides 

42 that are, for example, about 8 or more amino acids in length. Such fragments will typically 

43 comprise a domain or motif of the antigen peptide, e.g., active site, a transmembrane 

44 domain or a substrate-binding domain. Further, possible fragments include, but are not 

45 limited to, domain or motif containing fragments, soluble peptide fragments, and 

46 fragments containing immunogenic structures. Predicted domains and functional sites are 

47 readily identifiable by computer programs well known and readily available to those of skill 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



24 



(41860-189496) 



1 in the art (e.g., PROSITE analysis). The results of one such analysis are provided in FIG. 

2 2. 

3 

4 Polypeptides often contain amino acids other than the 20 amino acids commonly referred 

5 to as the 20 naturally occurring amino acids. Further, many amino acids, including the 

6 terminal amino acids, may be modified by natural processes, such as processing and 

7 other post-translational modifications, or by chemical modification techniques well known 

8 in the art. Common modifications that occur naturally in antigen peptides are described in 

9 basic texts, detailed monographs, and the research literature, and they are well known to 
10 those of skill in the art. 

11 

12 Known modifications include, but are not limited to, acetylation, acylation, ADP- 

13 ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme 

14 moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment 

15 of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, 

16 cyclization, disulfide bond formation, demethylation, formation of covalent crosslinks, 

17 formation of cystine, formation of pyroglutamate, formylation, gamma carboxylation, 

18 glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, 

19 oxidation, proteolytic processing, phosphorylation, prenylation, racemization, 

20 selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such 

21 as arginylation, and ubiquitination. 
22 

23 Such modifications are well known to those of skill in the art and have been described in 

24 great detail in the scientific literature. Several particularly common modifications, 

25 glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, 

26 hydroxylation and ADP-ribosylation, for instance, are described in most basic texts, such 

27 as Proteins-Structure and Molecular Properties, 2.sup.nd Ed., T. E. Creighton, W. H. 

28 Freeman and Company, New York (1993). Many detailed reviews are available on this 

29 subject, such as by Wold, F., Posttranslational Covalent Modification of Proteins, B. C. 

30 Johnson, Ed., Academic Press, New York 1-12 (1983); Seifter et al. (Meth. Enzymol. 182: 

31 626-646 (1990)) and Rattan et al. (Ann. N.Y Acad. Sci. 663:48-62 (1992)). 
32 

33 Accordingly, the antigen peptides of the present invention also encompass derivatives or 

34 analogs in which a substituted amino acid residue is not one encoded by the genetic 

35 code, in which a substituent group is included, in which the mature antigen peptide is 

36 fused with another compound, such as a compound to increase the half-life of the antigen 

37 peptide (for example, polyethylene glycol), or in which the additional amino acids are 

38 fused to the mature antigen peptide, such as a leader or secretory sequence or a 

39 sequence for purification of the mature antigen peptide or a pro-protein sequence. 
40 

41 Antibody Production 
42 

43 Although an amino acid sequence or oligopeptide used for antibody induction does not 

44 require biological activity, it must be immunogenic. A peptide, polypeptide, or protein used 

45 to induce specific antibodies may have an amino acid sequence consisting of at least five 

46 amino acids and preferably at least 1 0 amino acids. Short stretches of amino acid 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



25 



(41860-189496) 



1 sequence may be genetically or chemically fused with those of another protein such as 

2 keyhole limpet hemocyanin, and the chimeric peptide used for antibody production. 

3 Alternatively, the oligopeptide may be of sufficient length to contain an entire domain. 

4 

5 Antibodies specific for peptides, polypeptides, or proteins may be produced by inoculation 

6 of an appropriate animal with an antigenic fragment of the peptide, polypeptide, or 

7 protein. Antibody production includes not only the stimulation of an immune response by 

8 injection into animals, but also analogous processes such as the production of synthetic 

9 antibodies, the screening of recombinant immunoglobulin libraries for specific-binding 

10 molecules (Orlandi R, et al. [1989] PNAS 86:3833-3837, or Huse W. D. et al. [1989] 

11 Science 256:1275-1281), or the in vitro stimulation of lymphocyte populations. Current 

12 technology (Winter G. and Milstein C. [1991] Nature 349:293-299) provides for a number 

13 of highly specific binding reagents based on the principles of antibody formation. These 

14 techniques may be adapted to produce molecules which specifically bind antigen 

15 peptides. Antibodies or other appropriate molecules generated against a specific 

16 immunogenic peptide fragment or oligopeptide can be used in Western analysis, enzyme- 

17 linked immunosorbent assays (ELISA) or similar tests to establish the presence of or to 

18 quantitate amounts of peptide, polypeptide, or protein in normal, diseased, or transformed 

19 cells, tissues, organs, or organisms as well as liquid suspensions containing said peptide, 

20 polypeptide, or protein. 
21 

22 The invention also provides antibodies that selectively bind to one of the peptides of the 

23 present invention, a protein comprising such a peptide, as well as variants and fragments 

24 thereof. As used herein, an antibody selectively binds a target peptide when it binds the 

25 target peptide and does not significantly bind to unrelated proteins. An antibody is still 

26 considered to selectively bind a peptide even if it also binds to other proteins that are not 

27 substantially homologous with the target peptide so long as such proteins share 

28 homology with a fragment or domain of the peptide target of the antibody. In this case, it 

29 would be understood that antibody binding to the peptide is still selective despite some 

30 degree of cross-reactivity. 
31 

32 As used herein, an antibody is defined in terms consistent with that recognized within the 

33 art: they are multi-subunit proteins produced by a mammalian organism in response to an 

34 antigen challenge. The antibodies of the present invention include polyclonal antibodies 

35 and monoclonal antibodies, as well as fragments of such antibodies, including, but not 

36 limited to, Fab or F(ab , ).sub.2, and Fv fragments. 
37 

38 Many methods are known for generating and/or identifying antibodies to a given target 

39 peptide. Several such methods are described by Harlow, Antibodies, Cold Spring Harbor 

40 Press, (1989). 
41 

42 In general, to generate antibodies, an isolated peptide is used as an immunogen and is 

43 administered to a mammalian organism, such as a rat, rabbit or mouse. The full-length 

44 protein, an antigenic peptide fragment or a fusion protein can be used. Particularly 

45 important fragments are those covering functional domains, such as the domains 

46 identified in the figures, and domain of sequence homology or divergence amongst the 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



26 



(41860-189496) 



1 family, such as those that can readily be identified using protein alignment methods and 

2 as presented in the Figures. 

3 

4 Antibodies are preferably prepared from regions or discrete fragments of the antigen 

5 proteins. Antibodies can be prepared from any region of the peptide as described herein. 

6 However, preferred regions will include those involved in function/activity and/or 

7 antigen/binding partner interaction. 

8 

9 An antigenic fragment will typically comprise at least 8 contiguous amino acid residues. 

10 The antigenic peptide can comprise, however, at least 10, 12, 14, 16 or more amino acid 

1 1 residues. Such fragments can be selected on a physical property, such as fragments 

12 correspond to regions that are located on the surface of the protein, e.g., hydrophilic 

13 regions or can be selected based on sequence uniqueness. 
14 

15 Detection on an antibody of the present invention can be facilitated by coupling (i.e., 

16 physically linking) the antibody to a detectable substance. Examples of detectable 

17 substances include various enzymes, prosthetic groups, fluorescent materials, 

18 luminescent materials, bioluminescent materials, and radioactive materials. Examples of 

19 suitable enzymes include horseradish peroxidase, alkaline phosphatase, .beta.- 

20 galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes 

21 include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials 

22 include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, 

23 dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a 

24 luminescent material includes luminol; examples of bioluminescent materials include 

25 luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 

26 .sup.1251, .sup.1311, .sup.35S or .sup.3H. 
27 

28 The antibodies can be used to isolate one of the proteins of the present invention by 

29 standard techniques, such as affinity chromatography or immunoprecipitation. The 

30 antibodies can facilitate the purification of the natural protein from cells and recombinantly 

31 produced protein expressed in host cells. In addition, such antibodies are useful to detect 

32 the presence of one of the proteins of the present invention in cells or tissues to 

33 determine the pattern of expression of the protein among various tissues in an organism 

34 and over the course of schizont development. Such antibodies can be used to detect 

35 protein in situ, in vitro, or in a cell lysate or supernatant in order to evaluate the 

36 abundance and pattern of expression. Also, such antibodies can be used to assess 

37 abnormal tissue distribution or abnormal expression during development or progression of 

38 a protozoan infection. Antibody detection of circulating fragments of the full length protein 

39 can be used to identify turnover. 
40 

41 Further, the antibodies can be used to assess expression in disease states such as in 

42 active stages of the disease or in lymphocytes harvested from infected animals. 

43 Experimental data as provided in FIG. 22 indicates expression in bovine lymphocytes, 

44 infected with T. parva. If a disease is characterized by a specific mutation in the protein, 

45 antibodies specific for this mutant protein can be used to assay for the presence of the 

46 specific mutant protein. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



27 



(41860-189496) 



1 

2 The diagnostic uses can be applied, not only in genetic testing, but also in monitoring a 

3 treatment modality. Accordingly, where treatment is ultimately aimed at preventing or 

4 controlling infection, antibodies directed against the protein or relevant fragments can be 

5 used to monitor therapeutic efficacy. 
6 

7 The antibodies are also useful for inhibiting protein function, for example, blocking the 

8 binding of the antigenic peptide to a binding partner such as a substrate. These uses can 

9 also be applied in a therapeutic context in which treatment involves inhibiting the protein's 

10 function. An antibody can be used, for example, to block binding, thus modulating 

1 1 (agonizing or antagonizing) the peptides activity. Antibodies can be prepared against 

12 specific fragments containing sites required for function or against intact protein that is 

13 associated with a cell or cell membrane. See FIGS. 16-20 or 23, for structural information 

14 relating to the proteins of the present invention. 
15 

16 The invention also encompasses kits for using antibodies to detect the presence of a 

17 protein in a biological sample. The kit can comprise antibodies such as a labeled or 

18 labelable antibody and a compound or agent for detecting protein in a biological sample; 

19 means for determining the amount of protein in the sample; means for comparing the 

20 amount of protein in the sample with a standard; and instructions for use. Such a kit can 

21 be supplied to detect a single protein or epitope or can be configured to detect one of a 

22 multitude of epitopes, such as in an antibody detection array. Arrays are described in 

23 detail below for nuleic acid arrays and similar methods have been developed for antibody 

24 arrays. 
25 

26 Nucleic Acid Molecules 

27 

28 The present invention further provides isolated nucleic acid molecules that encode a T. 

29 parva antigen peptide or protein of the present invention (cDNA, transcript and genomic 

30 sequence). Such nucleic acid molecules will consist of, consist essentially of, or comprise 

31 a nucleotide sequence that encodes one of the enzyme peptides of the present invention, 

32 an allelic variant thereof, or an ortholog or paralog thereof. 
33 

34 As used herein, an "isolated" nucleic acid molecule is one that is separated from other 

35 nucleic acid present in the natural source of the nucleic acid. Preferably, an "isolated" 

36 nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences 

37 located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from 

38 which the nucleic acid is derived. However, there can be some flanking nucleotide 

39 sequences, for example up to about 5KB, 4KB, 3KB, 2KB, or 1 KB or less, particularly 

40 contiguous peptide encoding sequences and peptide encoding sequences within the 

41 same gene but separated by introns in the genomic sequence. The important point is that 

42 the nucleic acid is isolated from remote and unimportant flanking sequences such that it 

43 can be subjected to the specific manipulations described herein such as recombinant 

44 expression, preparation of probes and primers, and other uses specific to the nucleic acid 

45 sequences. 
46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



28 



(41860-189496) 



1 Moreover, an "isolated" nucleic acid molecule, such as a transcript/cDNA molecule, can 

2 be substantially free of other cellular material, or culture medium when produced by 

3 recombinant techniques, or chemical precursors or other chemicals when chemically 

4 synthesized. However, the nucleic acid molecule can be fused to other coding or 

5 regulatory sequences and still be considered isolated. 
6 

7 For example, recombinant DNA molecules contained in a vector are considered isolated. 

8 Further examples of isolated DNA molecules include recombinant DNA molecules 

9 maintained in heterologous host cells or purified (partially or substantially) DNA molecules 

10 in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the 

11 isolated DNA molecules of the present invention. Isolated nucleic acid molecules 

12 according to the present invention further include such molecules produced synthetically. 
13 

14 Following are examples which illustrate procedures for practicing the invention. These 

15 examples should not be construed as limiting. All percentages are by weight and all 

16 solvent mixture proportions are by volume unless otherwise noted. As used herein and 

17 further defined below, "nucleic acid" may refer to either DNA or RNA, or molecules which 

18 contain both deoxy- and ribonucleotides. The nucleic acids include genomic DNA, cDNA, 

19 and oligonucleotides including sense and anti-sense nucleic acids. Such nucleic acids 

20 may also contain modifications in the ribose-phosphate backbone to increase stability and 

21 half life of such molecules in physiological environments. 
22 

23 The nucleic acid may be double stranded, single stranded, or contain portions of both 

24 double stranded or single stranded sequence. As will be appreciated by those in the art, 

25 the depiction of a single strand ('Watson") also defines the sequence of the other strand 

26 ("Crick"); thus the sequences depicted in the figures also include the complement of the 

27 sequence. 
28 

29 One method for controlling gene expression according to the subject invention provides 

30 materials that would inform the production of double-stranded interfering RNA (dsRNAi), 

31 or RNA-mediated interference (RNAi), using published methods. The terms dsRNAi and 

32 RNAi are used interchangeably herein unless otherwise noted. In a more preferred 

33 embodiment, compositions are useful for regulation of gene expression in bovine 

34 lymphocytes. Such RNA would contain a nucleotide sequence identical to a fragment of 

35 the target gene; however, RNA sequences with insertions, deletions, and point mutations 

36 relative to the target sequence can also be used for inhibition. Sequence identity may 

37 optimized by sequence comparison and alignment algorithms known in the art (see 

38 Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991 , and 

39 references cited therein) and calculating the percent difference between the nucleotide 

40 sequences by, for example, the Smith-Waterman algorithm as implemented in the 

41 BESTFIT software program using default parameters (e.g., University of Wisconsin 

42 Genetic Computing Group). Alternatively, the duplex region of the RNA may be defined 

43 functionally as a nucleotide sequence that is capable of hybridizing with a fragment of the 

44 target gene transcript. 
45 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



29 



(41860-189496) 



1 As disclosed herein, 100% sequence identity between the RNA and the target gene is not 

2 required to practice the present invention. Thus the invention has the advantage of being 

3 able to tolerate sequence variations that might be expected due to genetic mutation, 

4 strain polymorphism, or evolutionary divergence. RNAi molecules of the subject invention 

5 are not limited to those that are targeted to the full-length polynucleotide or gene. Gene 

6 product can be inhibited with a RNAi molecule that is targeted to a portion or fragment of 

7 the exemplified polynucleotides; high homology (90-95%) or greater identity is also 

8 preferred, but not necessarily essential, for such applications. 
9 

10 

11 Protein/Peptide Uses 
12 

13 The proteins of the present invention can be used in substantial and specific assays 

14 related to the functional information provided in the Figures; for example, to stimulate 

15 CD8+ cytotoxic T cell lines, to raise antibodies or to elicit another immune response; as a 

16 reagent (including the labeled reagent) in assays designed to quantitatively determine 

17 levels of the protein (or its binding partner or ligand) in biological fluids; and as markers 

18 for tissues in which the corresponding protein is preferentially expressed (either 

19 constitutively or at a particular stage of tissue differentiation or development or in a 

20 disease state). Where the protein binds or potentially binds to another protein or ligand 

21 (such as, for example, in a enzyme-effector protein interaction or enzyme-ligand 

22 interaction), the protein can be used to identify the binding partner/ligand so as to develop 

23 a system to identify inhibitors of the binding interaction. Any or all of these uses are 

24 capable of being developed into reagent grade or kit format for commercialization as 

25 commercial products. 
26 

27 Methods for performing the uses listed above are well known to those skilled in the art. 

28 References disclosing such methods include "Molecular Cloning: A Laboratory Manual", 

29 2d ed., Cold Spring Harbor Laboratory Press, Sambrook, J., E. F. Fritsch and T. Maniatis 

30 eds., 1989, and "Methods in Enzymology: Guide to Molecular Cloning Techniques", 

31 Academic Press, Berger, S. L. and A. R. Kimmel eds., 1987. 
32 

33 The potential uses of the peptides of the present invention are based primarily on the 

34 source of the protein as well as the class/action of the protein. For example, antigens 

35 isolated from T. parva and their protozoan orthologs serve as targets for identifying 

36 agents for use in mammalian therapeutic applications, e.g. an animal drug, particularly in 

37 modulating a biological or pathological response in a cell, tissue, or protozoan, that 

38 expresses an immunogenic antigen. Experimental data as provided in FIG. 23 indicates 

39 that the antigens of the present invention are expressed in infected host lymphocytes, as 

40 indicated by flow cytometric analysis. Such uses can readily be determined using the 

41 information provided herein, that which is known in the art, and routine experimentation. 
42 

43 The proteins of the present invention are also useful in drug screening assays, in cell- 

44 based or cell-free systems. Cell-based systems can be native, i.e., cells that normally 

45 express the enzyme, as a biopsy or expanded in cell culture. Experimental data as 

46 provided in FIG. 22 indicates expression in infected bovine lymphocytes. In an alternate 

47 embodiment, cell-based assays involve recombinant host cells expressing the antigen 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



30 



(41860-189496) 



1 protein. 

2 

3 The polypeptides can be used to identify compounds that modulate pathogen activity as 

4 measured by the antigen in its natural state or an altered form that causes a specific 

5 disease or pathology associated with the protozoan. Both the antigens of the present 

6 invention and appropriate variants and fragments can be used in high-throughput screens 

7 to assay candidate compounds for the ability to bind to the antigen. These compounds 

8 can be further screened against a functional T-cell antigen to determine the effect of the 

9 compound on the enzyme activity. Further, these compounds can be tested in animal or 
10 invertebrate systems to determine activity/effectiveness. 

11 

12 The invention further includes other end point assays to identify compounds that 

13 modulate (stimulate or inhibit) protozoan antigen expression. 
14 

15 The proteins of the present invention are also useful in competition binding assays in 

16 methods designed to discover compounds that interact with the T. parva antigens (e.g. 

17 binding partners and/or ligands). Thus, a compound is exposed to an antigen polypeptide 

18 under conditions that allow the compound to bind or to otherwise interact with the 

19 polypeptide. Soluble antigen polypeptide is also added to the mixture. If the test 

20 compound interacts with the soluble antigen polypeptide, it decreases the amount of 

21 complex formed or activity from the antigen target. This type of assay is particularly useful 

22 in cases in which compounds are sought that interact with specific regions of the antigen. 

23 Thus, the soluble polypeptide that competes with the target antigen region is designed to 

24 contain peptide sequences corresponding to the region of interest. 
25 

26 To perform cell free drug screening assays, it is sometimes desirable to immobilize either 

27 the antigen protein, or fragment, or its target molecule to facilitate separation of 

28 complexes from uncomplexed forms of one or both of the proteins, as well as to 

29 accommodate automation of the assay. 

30 

31 Techniques for immobilizing proteins on matrices can be used in the drug screening 

32 assays. In one embodiment, a fusion protein can be,provided which adds a domain that 

33 allows the protein to be bound to a matrix. For example, glutathione-S-transferase fusion 

34 proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, 

35 Mo.) or glutathione derivatized microtitre plates, which are then combined with the cell 

36 lysates (e.g., .sup.35S-labeled) and the candidate compound, and the mixture incubated 

37 under conditions conducive to complex formation (e.g., at physiological conditions for salt 

38 and pH). Following incubation, the beads are washed to remove any unbound label, and 

39 the matrix immobilized and radiolabel determined directly, or in the supernatant after the 

40 complexes are dissociated. Alternatively, the complexes can be dissociated from the 

41 matrix, separated by SDS-PAGE, and the level of antigen-binding protein found in the 

42 bead fraction quantitated from the gel using standard electrophoretic techniques. For 

43 example, either the polypeptide or its target molecule can be immobilized utilizing 

44 conjugation of biotin and streptavidin using techniques well known in the art. Alternatively, 

45 antibodies reactive with the protein but which do not interfere with binding of the protein to 

46 its target molecule can be derivatized to the wells of the plate, and the protein trapped in 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



31 



(41860-189496) 



1 the wells by antibody conjugation. Preparations of an antigen-binding protein and a 

2 candidate compound are incubated in the antigen protein-presenting wells and the 

3 amount of complex trapped in the well can be quantitated. Methods for detecting such 

4 complexes, in addition to those described above for the GST-immobilized complexes, 

5 include immunodetection of complexes using antibodies reactive with the antigen protein 

6 target molecule, or which are reactive with antigen protein and compete with the target 

7 molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity 

8 associated with the presence of the target molecule. 
9 

10 Agents that modulate one of the antigens of the present invention can be identified using 

1 1 one or more of the above assays, alone or in combination. It is generally preferable to use 

12 a cell-based or cell free system first and then confirm activity in an animal or other model 

13 system. Such model systems are well known in the art and can readily be employed in 

14 this context. 
15 

16 

17 In yet another aspect of the invention, the antigen proteins can be used as "bait proteins" 

18 in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et 

19 al. (1993) Cell 72:223-232; Madura et al. (1993) J. Biol. Chem. 268:12046-12054; Bartel 

20 et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; 

21 and Brent WO94/10300), to identify other proteins, which bind to or interact with the 

22 antigen and are involved in T-cell induction activity. Such antigen-binding proteins are 

23 also likely to be involved in the propagation of signals by antigen-binding proteins or 

24 antigen targets as, for example, downstream elements of a MHC Class l-mediated 

25 signaling pathway. Alternatively, such antigen-binding proteins could act in a manner as 

26 to be inhibitors of an immune response. 
27 

28 

29 This invention further pertains to novel agents identified by the above-described 

30 screening assays. Accordingly, it is within the scope of this invention to further use an 

31 agent identified as described herein in an appropriate animal model. For example, an 

32 agent identified as described herein (e.g., an antigen-modulating agent, an antisense 

33 antigen nucleic acid molecule, or an antigen-specific antibody, can be used in an animal 

34 or other model to determine the efficacy, toxicity, or side effects of treatment with such an 

35 agent. Alternatively, an agent identified as described herein can be used in an animal or 

36 other model to determine the mechanism of action of such an agent. Furthermore, this 

37 invention pertains to uses of novel agents identified by the above-described screening 

38 assays for treatments as described herein. 
39 

40 The antigen proteins of the present invention are also useful to provide a target for 

41 diagnosing a disease or a disease mediated by the peptide. Accordingly, the invention 

42 provides methods for detecting the presence, or levels of, the protein (or encoding 

43 mRNA) in a cell, tissue, or organism. Experimental data as provided in FIG. 22 indicates 

44 expression in T. parva-infected bovine lymphocytes. The method involves contacting a 

45 biological sample with a compound capable of interacting with the antigen protein such 

46 that the interaction can be detected. Such an assay can be provided in a single detection 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



32 



(41860-189496) 



1 format or a multi-detection format such as an antibody chip array. 

2 

3 One agent for detecting a protein in a sample is an antibody capable of selectively binding 

4 to protein. A biological sample includes tissues, cells and biological fluids isolated from a 

5 subject, as well as tissues, cells and fluids present within a subject. 

6 

7 The peptides of the present invention also provide targets for diagnosing active parasite 

8 activity or disease, in an animal having a variant peptide, particularly activities and 

9 conditions that are known for other members of the family of proteins to which the present 

10 one belongs. Thus, the peptide can be isolated from a biological sample and assayed for 

11 the presence of a genetic mutation that results in aberrant peptide. This includes amino 

12 acid substitution, deletion, insertion, rearrangement, (as the result of aberrant splicing 

13 events), and inappropriate post-translational modification. Analytic methods include 

14 altered electrophoretic mobility, altered tryptic peptide digest, altered enzyme activity in 

15 cell-based or cell-free assay, alteration in substrate or antibody-binding pattern, altered 

16 isoelectric point, direct amino acid sequencing, and any other of the known assay 

17 techniques useful for detecting mutations in a protein. Such an assay can be provided in 

18 a single detection format or a multi-detection format such as an antibody chip array. 
19 

20 In vitro techniques for detection of peptide include enzyme linked immunosorbent assays 

21 (ELISAs), Western blots, immunoprecipitations and immunofluorescence using a 

22 detection reagent, such as an antibody or protein binding agent, as well as the methods 

23 that represent embodiments of the present invention. Alternatively, the peptide can be 

24 detected in vivo in a subject by introducing into the subject a labeled anti-peptide antibody 

25 or other types of detection agent. For example, the antibody can be labeled with a 

26 radioactive marker whose presence and location in a subject can be detected by standard 

27 imaging techniques. Such methods can be used to detect the allelic variant of a peptide 

28 expressed in an infected subject and methods which detect fragments of a peptide in a 

29 sample. 
30 

31 The diagnostic uses can be applied, not only in genetic testing, but also in monitoring a 

32 treatment modality. Accordingly, where treatment is ultimately aimed at preventing or 

33 controlling infection, antibodies directed against the protein or relevant fragments can be 

34 used to monitor therapeutic efficacy. 

35 

36 The antibodies are also useful for inhibiting protein function, for example, blocking the 

37 binding of the antigenic peptide to a binding partner such as a substrate. These uses can 

38 also be applied in a therapeutic context in which treatment involves inhibiting the protein's 

39 function. An antibody can be used, for example, to block binding, thus modulating 

40 (agonizing or antagonizing) the peptides activity. Antibodies can be prepared against 

41 specific fragments containing sites required for function or against intact protein that is 

42 associated with a cell or cell membrane. See FIGS. 1 6-20 and 23 for structural 

43 information relating to the proteins of the present invention. 
44 

45 The invention also encompasses kits for using antibodies to detect the presence of a 

46 protein in a biological sample. The kit can comprise antibodies such as a labeled or 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



33 



(41860-189496) 



1 labelable antibody and a compound or agent for detecting protein in a biological sample; 

2 means for determining the amount of protein in the sample; means for comparing the 

3 amount of protein in the sample with a standard; and instructions for use. Such a kit can 

4 be supplied to detect a single protein or epitope or can be configured to detect one of a 

5 multitude of epitopes, such as in an antibody detection array. Arrays are described in 

6 detail below for nucleic acid arrays and similar methods have been developed for 

7 antibody arrays. 
8 

9 Nucleic Acid Molecules 
10 

1 1 [0104] The present invention further provides isolated nucleic acid molecules that encode 

12 a T. parva antigen peptide or protein of the present invention (cDNA, transcript and 

13 genomic sequence). Such nucleic acid molecules will consist of, consist essentially of, or 

14 comprise a nucleotide sequence that encodes one of the enzyme peptides of the present 

15 invention, an allelic variant thereof, or an ortholog or paralog thereof. 
16 

17 As used herein, an "isolated" nucleic acid molecule is one that is separated from other 

18 nucleic acid present in the natural source of the nucleic acid. Preferably, an "isolated" 

19 nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences 

20 located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from 

21 which the nucleic acid is derived. However, there can be some flanking nucleotide 

22 sequences, for example up to about 5KB, 4KB, 3KB, 2KB, or 1KB or less, particularly 

23 contiguous peptide encoding sequences and peptide encoding sequences within the 

24 same gene but separated by introns in the genomic sequence. The important point is that 

25 the nucleic acid is isolated from remote and unimportant flanking sequences such that it 

26 can be subjected to the specific manipulations described herein such as recombinant 

27 expression, preparation of probes and primers, and other uses specific to the nucleic acid 

28 sequences. 
29 

30 Moreover, an "isolated" nucleic acid molecule, such as a transcript/cDNA molecule, can 

31 be substantially free of other cellular material, or culture medium when produced by 

32 recombinant techniques, or chemical precursors or other chemicals when chemically 

33 synthesized. However, the nucleic acid molecule can be fused to other coding or 

34 regulatory sequences and still be considered isolated. 
35 

36 For example, recombinant DNA molecules contained in a vector are considered isolated. 

37 Further examples of isolated DNA molecules include recombinant DNA molecules 

38 maintained in heterologous host cells or purified (partially or substantially) DNA molecules 

39 in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the 

40 isolated DNA molecules of the present invention. Isolated nucleic acid molecules 

41 according to the present invention further include such molecules produced synthetically. 
42 

43 Accordingly, the present invention provides nucleic acid molecules that consist of the 

44 nucleotide sequence shown in FIGS. 16-20 or any nucleic acid molecule that encodes the 

45 protein (amino acid sequence) included in these figures. A nucleic acid molecule consists 

46 of a nucleotide sequence when the nucleotide sequence is the complete nucleotide 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



34 



(41860-189496) 



1 sequence of the nucleic acid molecule. 

2 

3 The present invention further provides nucleic acid molecules that consist essentially of 

4 the nucleotide sequence shown in FIGS. 16-20, or any nucleic acid molecule that 

5 encodes the proteins provided in the same figures. A nucleic acid molecule consists 

6 essentially of a nucleotide sequence when such a nucleotide sequence is present with 

7 only a few additional nucleic acid residues in the final nucleic acid molecule. 
8 

9 The present invention further provides nucleic acid molecules that comprise the 

10 nucleotide sequences shown shown in FIGS. 16-20, or any nucleic acid molecule that 

1 1 encodes the proteins provided in the same figures A nucleic acid molecule comprises a 

12 nucleotide sequence when the nucleotide sequence is at least part of the final nucleotide 

13 sequence of the nucleic acid molecule. In such a fashion, the nucleic acid molecule can 

14 be only the nucleotide sequence or have additional nucleic acid residues, such as nucleic 

15 acid residues that are naturally associated with it or heterologous nucleotide sequences. 

16 Such a nucleic acid molecule can have a few additional nucleotides or can comprises 

17 several hundred or more additional nucleotides. A brief description of how various types 

18 of these nucleic acid molecules can be readily made/isolated is provided below. 
19 

20 

21 The invention further provides nucleic acid molecules that encode fragments of the 

22 peptides of the present invention as well as nucleic acid molecules that encode obvious 

23 variants of the enzyme proteins of the present invention that are described above. Such 

24 nucleic acid molecules may be naturally occurring, such as allelic variants (same locus), 

25 paralogs (different locus), and orthologs (different organism), or may be constructed by 

26 recombinant DNA methods or by chemical synthesis. Such non-naturally occurring 

27 variants may be made by mutagenesis techniques, including those applied to nucleic acid 

28 molecules, cells, or organisms. Accordingly, as discussed above, the variants can contain 

29 nucleotide substitutions, deletions, inversions and insertions. Variation can occur in either 

30 or both the coding and non-coding regions. The variations can produce both conservative 

31 and non- conservative amino acid substitutions. 
32 

33 

34 The present invention further provides epitope fragments of the antigen proteins and the 

35 nucleic acid molecules encoding the antigens, provided in the description of epitope- 

36 mapping experiments described below. An epitope coding region comprises a contiguous 

37 nucleotide sequence greater than 12 or more nucleotides. Further, a fragment could at 

38 least 30, 40, 50, 100, 250 or 500 nucleotides in length. The length of the fragment will be 

39 based on its intended use. For example, the fragment can encode epitope bearing 

40 regions of the peptide, or can be useful as DNA probes and primers. Such fragments can 

41 be isolated using the known nucleotide sequence to synthesize an oligonucleotide probe. 

42 A labeled probe can then be used to screen a cDNA library, genomic DNA library, or 

43 mRNA to isolate nucleic acid corresponding to the coding region. Further, primers can be 

44 used in PCR reactions to clone specific regions of gene. 
45 

46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



35 



(41860-189496) 



1 A probe/primer typically comprises substantially a purified oligonucleotide or 

2 oligonucleotide pair. The oligonucleotide typically comprises a region of nucleotide 

3 sequence that hybridizes under stringent conditions to at least about 12, 20, 25, 40, 50 or 

4 more consecutive nucleotides. 

5 
6 
7 
8 
9 
10 

11 Orthologs, homologs, and allelic variants can be identified using methods well known in 

12 the art. As described in the Peptide Section, these variants comprise a nucleotide 

13 sequence encoding a peptide that is typically 60-70%, 70-80%, 80-90%, and more 

14 typically at least about 90-95% or more homologous to the nucleotide sequence shown in 

15 the Figure sheets or a fragment of this sequence. Such nucleic acid molecules can readily 

16 be identified as being able to hybridize under moderate to stringent conditions, to the 

17 nucleotide sequence shown in the Figure sheets or a fragment of the sequence. Allelic 

18 variants can readily be determined by genetic locus of the encoding gene. The gene 

19 encoding the novel enzyme of the present invention is located on a genome component 

20 that has been mapped to the T. parva chromosomel and 2 which is supported by multiple 

21 lines of evidence, such as STS and BAC map data. 
22 

23 

24 Nucleic Acid Molecule Uses 

25 

26 The nucleic acid molecules of the present invention are useful for probes, primers, 

27 chemical intermediates, and in biological assays. The nucleic acid molecules are useful 

28 as a hybridization probe for messenger RNA, transcript/cDNA and genomic DNA to 

29 isolate full-length cDNA and genomic clones encoding the peptide described in FIG. 16- 

30 20, and to isolate cDNA and genomic clones that correspond to variants (alleles, 

31 orthologs, etc.) producing the same or related peptides shown in FIG. 16-20 and FIG 23. 

32 As illustrated in FIG. 1 1 , deletions were identified at 2 different nucleotide positions. 
33 

34 The probe can correspond to any sequence along the entire length of the nucleic acid 

35 molecules provided in the Figures. Accordingly, it could be derived from 5' noncoding 

36 regions, the coding region, and 3' noncoding regions. However, as discussed, fragments 

37 are not to be construed as encompassing fragments disclosed prior to the present 

38 invention. 
39 

40 The nucleic acid molecules are also useful as primers for PCR to amplify any given region 

41 of a nucleic acid molecule and are useful to synthesize antisense molecules of desired 

42 length and sequence. 

43 

44 The nucleic acid molecules are also useful for constructing recombinant vectors. Such 

45 vectors include expression vectors that express a portion of, or all of, the peptide 

46 sequences. Vectors also include insertion vectors, used to integrate into another nucleic 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



36 



(41860-189496) 



1 acid molecule sequence, such as into the cellular genome, to alter in situ expression of a 

2 gene and/or gene product. For example, an endogenous coding sequence can be 

3 replaced via homologous recombination with all or part of the coding region containing 

4 one or more specifically introduced mutations. 

5 
6 

7 The nucleic acid molecules are also useful for expressing antigenic portions of the 

8 proteins. 

9 

10 The nucleic acid molecules are also useful as probes for determining the chromosomal 

1 1 positions of the nucleic acid molecules by means of in situ hybridization methods. The 

12 genes encoding the novel antigens of the present invention are located on a genome 

13 component that has been mapped to T. pan/a chromosomes 1 (Tp1 & Tp4) and 2 (Tp5, 

14 Tp7&Tp8). 
15 

16 The nucleic acid molecules are also useful in making vectors containing the gene 

17 regulatory regions of the nucleic acid molecules of the present invention. 

18 

19 The nucleic acid molecules are also useful for designing ribozymes/antigens 

20 corresponding to all, or a part, of the mRNA produced from the nucleic acid molecules 

21 described herein. 

22 

23 The nucleic acid molecules are also useful for making vectors that express part, or all, of 

24 the peptides. 

25 

26 The nucleic acid molecules are also useful for constructing host cells expressing a part, or 

27 all, of the nucleic acid molecules and peptides. 

28 

29 The nucleic acid molecules are also useful for constructing transgenic animals expressing 

30 all, or a part, of the nucleic acid molecules and peptides. 

31 

32 The nucleic acid molecules are also useful as hybridization probes for determining the 

33 presence, level, form and distribution of nucleic acid expression. Such probes could be 

34 used to detect the presence of, or to determine levels of, a specific nucleic acid molecule 

35 in cells, tissues, and in organisms. The nucleic acid whose level is determined can be 

36 DNA or RNA. Accordingly, probes corresponding to the peptides described herein can be 

37 used to assess expression and/or gene copy number in a given cell, tissue, or organism. 

38 These uses are relevant for diagnosis of disorders involving an increase or decrease in 

39 antigen protein expression relative to normal results. 
40 

41 In vitro techniques for detection of mRNA include Northern hybridizations and in situ 

42 hybridizations. In vitro techniques for detecting DNA includes Southern hybridizations and 

43 in situ hybridization. 
44 

45 Probes can be used as a part of a diagnostic test kit for identifying cells or tissues that 

46 express an T. parva antigen protein, such as by measuring a level of an antigen-encoding 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



37 



(41860-189496) 



1 nucleic acid in a sample of cells from a subject e.g., mRNA or genomic DNA, or 

2 determining if an antigen gene has been mutated. 

3 

4 Nucleic acid expression assays are useful for drug screening to identify compounds that 

5 modulate protozoan antigen nucleic acid expression. 

6 

7 The invention thus provides a method for identifying a compound that can be used to treat 

8 a disease associated with nucleic acid expression of the antigen gene, particularly 

9 biological and pathological processes that are mediated by the presence of T. parva in 

10 cells and tissues that express it. The method typically includes assaying the ability of the 

1 1 compound to modulate the expression of the antigen nucleic acid and thus identifying a 

12 compound that can be used to treat a disorder characterized by nucleic acid expression 

13 by an infecting protozoan. The assays can be performed in cell-based and cell-free 

14 systems. Cell-based assays include cells naturally expressing the enzyme nucleic acid or 

15 recombinant cells genetically engineered to express specific nucleic acid sequences. 

16 

17 The assay for enzyme nucleic acid expression can involve direct assay of nucleic acid 

18 levels, such as mRNA levels, or on collateral compounds involved in an antigen-specific 

19 signal pathway. Further, the expression of genes that are up- or down-regulated in 

20 response to the antigen-stimulating signal pathway can also be assayed. In this 

21 embodiment the regulatory regions of these genes can be operably linked to a reporter 

22 gene such as luciferase. 
23 

24 Thus, modulators of antigen gene expression can be identified in a method wherein a cell 

25 is contacted with a candidate compound and the expression of mRNA determined. The 

26 level of expression of antigen mRNA in the presence of the candidate compound is 

27 compared to the level of expression of antigen mRNA in the absence of the candidate 

28 compound. The candidate compound can then be identified as a modulator of nucleic 

29 acid expression based on this comparison and be used, for example to treat an infection 

30 by a pathogenic protozoan. When expression of mRNA is statistically significantly greater 

31 in the presence of the candidate compound than in its absence, the candidate compound 

32 is identified as a stimulator of nucleic acid expression. When nucleic acid expression is 

33 statistically significantly less in the presence of the candidate compound than in its 

34 absence, the candidate compound is identified as an inhibitor of nucleic acid expression. 
35 

36 

37 The nucleic acid molecules are also useful for monitoring the effectiveness of modulating 

38 compounds on the infectivity of a protozoan in clinical trials or in a treatment regimen. 

39 Thus, the gene expression pattern can serve as a barometer for the continuing 

40 effectiveness of treatment with the compound, particularly with compounds to which an 

41 animal or the pathogen can develop resistance. The gene expression pattern can also 

42 serve as a marker indicative of a physiological response of the affected cells to the 

43 compound. 
44 

45 Accordingly, such monitoring would allow either increased administration of the 

46 compound or the administration of alternative compounds to which the parasite has not 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



38 



(41860-189496) 



1 become resistant. Similarly, if the level of nucleic acid expression falls below a desirable 

2 level, administration of the compound could be commensurately decreased. 

3 

4 The nucleic acid molecules are also useful in diagnostic assays for qualitative changes in 

5 antigen nucleic acid expression, and particularly in qualitative changes that lead to 

6 pathology. The nucleic acid molecules can be used to detect mutations in antigen genes 

7 and gene expression products such as mRNA. The nucleic acid molecules can be used 

8 as hybridization probes to detect naturally occurring genetic mutations in the antigen gene 

9 and thereby to determine whether a parasite with the mutation is of a different strain or 

10 has a differential pathogenic potential. Mutations include deletion, addition, or substitution 

11 of one or more nucleotides in the gene, chromosomal rearrangement, such as inversion 

12 or transposition, modification of genomic DNA, such as aberrant methylation patterns or 

13 changes in gene copy number, such as amplification. Detection of a mutated form of the 

14 antigen gene associated with a dysfunction provides a diagnostic tool for an active 

15 disease or susceptibility to disease when the disease results from over-expression, 

16 under-expression, or altered expression of an antigen protein. 
17 

18 Parasite strains carrying mutations in the enzyme gene can be detected at the nucleic 

19 acid level by a variety of techniques. SNPs that have been found in the gene encoding 

20 the antigen of the present invention. SNPs were identified at 4 different nucleotide 

21 positions. Genomic DNA can be analyzed directly or can be amplified by using PCR prior 

22 to analysis. RNA or cDNA can be used in the same way. In some uses, detection of the 

23 mutation involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, 

24 e.g. U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, 

25 alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al., Science 

26 241:1077-1080 (1988); and Nakazawa et al., PNAS 91:360-364 (1994)), the latter of 

27 which can be particularly useful for detecting point mutations in the gene (see Abravaya 

28 et al., Nucleic Acids Res. 23:675-682 (1995)). This method can include the steps of 

29 collecting a sample of cells from an infective parasite, isolating nucleic acid (e.g., 

30 genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample 

31 with one or more primers which specifically hybridize to a gene under conditions such that 

32 hybridization and amplification of the gene (if present) occurs, and detecting the presence 

33 or absence of an amplification product, or detecting the size of the amplification product 

34 and comparing the length to a control sample. Deletions and insertions can be detected 

35 by a change in size of the amplified product compared to the normal genotype. Point 

36 mutations can be identified by hybridizing amplified DNA to normal RNA or antisense 

37 DNA sequences. 
38 

39 Alternatively, mutations in a enzyme gene can be directly identified, for example, by 

40 alterations in restriction enzyme digestion patterns determined by gel electrophoresis. 
41 

42 Further, sequence-specific ribozymes (U.S. Pat. No. 5,498,531) can be used to score for 

43 the presence of specific mutations by development or loss of a ribozyme cleavage site. 

44 Perfectly matched sequences can be distinguished from mismatched sequences by 

45 nuclease cleavage digestion assays or by differences in melting temperature. 

46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



39 



(41860-189496) 



1 Sequence changes at specific locations can also be assessed by nuclease protection 

2 assays such as RNase and S1 protection or the chemical cleavage method. Furthermore, 

3 sequence differences between a mutant enzyme gene and a wild-type gene can be 

4 determined by direct DNA sequencing. A variety of automated sequencing procedures 

5 can be utilized when performing the diagnostic assays (Naeve, C. W., (1995) 

6 Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT 

7 International Publication No. WO94/16101 ; Cohen et al., Adv. Chromatogr. 36:127-162 

8 (1996); and Griffin et al., Appl. Biochem. Biotechnol. 38:147-159 (1993)). 
9 

10 Other methods for detecting mutations in the gene include methods in which protection 

1 1 from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA 

12 duplexes (Myers et al., Science 230:1242 (1985)); Cotton et al, PNAS 85:4397 (1988); 

13 Saleeba et al., Meth. Enzymol. 217:286-295 (1992)), electrophoretic mobility of mutant 

14 and wild type nucleic acid is compared (Orita et al., PNAS 86:2766 (1989); Cotton et al., 

15 Mutat. Res. 285:125-144 (1993); and Hayashi et al., Genet. Anal. Tech. Appl. 9:73-79 

16 (1992)), and movement of mutant or wild-type fragments in polyacryl amide gels 

17 containing a gradient of denaturant is assayed using denaturing gradient gel 

18 electrophoresis (Myers et al., Nature 313:495 (1985)). Examples of other techniques for 

19 detecting point mutations include selective oligonucleotide hybridization, selective 

20 amplification, and selective primer extension. 
21 

22 The nucleic acid molecules are also useful for testing an individual parasite for a 

23 genotype that while not necessarily causing the disease, nevertheless affects the 

24 treatment modality. Thus, the nucleic acid molecules can be used to study the 

25 relationship between a particular strain of parasite's genotype and the parasite's response 

26 to a compound used for treatment (pharmacogenomic relationship). Accordingly, the 

27 nucleic acid molecules described herein can be used to assess the mutation content of 

28 the antigen gene in an individual in order to select an appropriate compound or dosage 

29 regimen for treatment. FIG. 23 provides information on SNPs that have been found in 

30 eleven strains in the gene encoding the antigen of the present invention. SNPs were 

31 identified at nucleotide positions underlined in FIG 23. Some of these SNPs that are 

32 located outside the ORF and in introns may affect gene transcription. 
33 

34 Thus nucleic acid molecules displaying genetic variations that affect treatment provide a 

35 diagnostic target that can be used to tailor treatment in an individual. Accordingly, the 

36 production of recombinant cells and animals containing these polymorphisms allow 

37 effective clinical design of treatment compounds and dosage regimens. 
38 

39 The nucleic acid molecules are thus useful as antisense constructs to control antigen 

40 gene expression in cells, tissues, and organisms. A DNA antisense nucleic acid molecule 

41 is designed to be complementary to a region of the gene involved in transcription, 

42 preventing transcription and hence production of enzyme protein. An antisense RNA or 

43 DNA nucleic acid molecule would hybridize to the mRNA and thus block translation of 

44 mRNA into antigen protein. Alternatively, a class of antisense molecules can be used to 

45 inactivate mRNA in order to decrease expression of antigen nucleic acid. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



40 



(41860-189496) 



1 
2 

3 The invention also encompasses kits for detecting the presence of an antigen nucleic acid 

4 in a biological sample. For example, the kit can comprise reagents such as a labeled or 

5 labelable nucleic acid or agent capable of detecting antigen nucleic acid in a biological 

6 sample; means for determining the amount of antigen nucleic acid in the sample; and 

7 means for comparing the amount of antigen nucleic acid in the sample with a standard. 

8 The compound or agent can be packaged in a suitable container. The kit can further 

9 comprise instructions for using the kit to detect antigen protein mRNA or DNA. 
10 

11 Nucleic Acid Arrays 
12 

13 The present invention further provides nucleic acid detection kits, such as arrays or 

14 microarrays of nucleic acid molecules that are based on the sequence information 

15 provided in FIGS. 16-20. 
16 

17 As used herein "Arrays" or "Microarrays" refers to an array of distinct polynucleotides or 

18 oligonucleotides synthesized on a substrate, such as paper, nylon or other type of 

19 membrane, filter, chip, glass slide, or any other suitable solid support. In one 

20 embodiment, the microarray is prepared and used according to the methods described in 

21 U.S. Pat. No. 5,837,832, Chee et al., PCT application W095/1 1995 (Chee et al.), 

22 Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; 

23 Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their 

24 entirety by reference. In other embodiments, such arrays are produced by the methods 

25 described by Brown et al., U.S. Pat. No. 5,807,522. 
26 

27 The microarray or detection kit is preferably composed of a large number of unique, 

28 single-stranded nucleic acid sequences, usually either synthetic antisense 

29 oligonucleotides or fragments of cDNAs, fixed to a solid support. The oligonucleotides are 

30 preferably about 6-60 nucleotides in length, more preferably 15-30 nucleotides in length, 

31 and most preferably about 20-25 nucleotides in length. For a certain type of microarray or 

32 detection kit, it may be preferable to use oligonucleotides that are only 7-20 nucleotides in 

33 length. The microarray or detection kit may contain oligonucleotides that cover the known 

34 5\ or 3\ sequence, sequential oligonucleotides which cover the full length sequence; or 

35 unique oligonucleotides selected from particular areas along the length of the sequence. 

36 Polynucleotides used in the microarray or detection kit may be oligonucleotides that are 

37 specific to a gene or genes of interest. 
38 

39 In order to produce oligonucleotides to a known sequence for a microarray or detection 

40 kit, the gene(s) of interest (or an ORF identified in the present invention) is typically 

41 examined using a computer algorithm which starts at the 5' or at the 3' end of the 

42 nucleotide sequence. Typical algorithms will then identify oligomers of defined length that 

43 are unique to the gene, have a GC content within a range suitable for hybridization, and 

44 lack predicted secondary structure that may interfere with hybridization. In certain 

45 situations it may be appropriate to use pairs of oligonucleotides on a microarray or 

46 detection kit. The "pairs" will be identical, except for one nucleotide that preferably is 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



41 



(41860-189496) 



1 located in the center of the sequence. The second oligonucleotide in the pair 

2 (mismatched by one) serves as a control. The number of oligonucleotide pairs may range 

3 from two to one million. The oligomers are synthesized at designated areas on a 

4 substrate using a light-directed chemical process. The substrate may be paper, nylon or 

5 other type of membrane, filter, chip, glass slide or any other suitable solid support. 
6 

7 In another aspect, an oligonucleotide may be synthesized on the surface of the substrate 

8 by using a chemical coupling procedure and an ink jet application apparatus, as 

9 described in PCT application W095/251 116 (Baldeschweiler et al.) which is incorporated 

10 herein in its entirety by reference. In another aspect, a "gridded" array analogous to a dot 

1 1 (or slot) blot may be used to arrange and link cDNA fragments or oligonucleotides to the 

12 surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical 

13 bonding procedures. An array, such as those described above, may be produced by hand 

14 or by using available devices (slot blot or dot blot apparatus), materials (any suitable solid 

15 support), and machines (including robotic instruments), and may contain 8, 24, 96, 384, 

16 1 536, 61 44 or more oligonucleotides, or any other number between two and one million 

17 which lends itself to the efficient use of commercially available instrumentation. 
18 

19 In order to conduct sample analysis using a microarray or detection kit, the RNA or DNA 

20 from a biological sample is made into hybridization probes. The mRNA is isolated, and 

21 cDNA is produced and used as a template to make antisense RNA (aRNA). The aRNA is 

22 amplified in the presence of fluorescent nucleotides, and labeled probes are incubated 

23 with the microarray or detection kit so that the probe sequences hybridize to 

24 complementary oligonucleotides of the microarray or detection kit. Incubation conditions 

25 are adjusted so that hybridization occurs with precise complementary matches or with 

26 various degrees of less complementarity. After removal of nonhybridized probes, a 

27 scanner is used to determine the levels and patterns of fluorescence. The scanned 

28 images are examined to determine degree of complementarity and the relative 

29 abundance of each oligonucleotide sequence on the microarray or detection kit. The 

30 biological samples may be obtained from any bodily fluids (such as blood, urine, saliva, 

31 phlegm, gastric juices, etc.), cultured cells, biopsies, or other tissue preparations. A 

32 detection system may be used to measure the absence, presence, and amount of 

33 hybridization for all of the distinct sequences simultaneously. This data may be used for 

34 large-scale correlation studies on the sequences, expression patterns, mutations, 

35 variants, or polymorphisms among samples. 
36 

37 Using such arrays, the present invention provides methods to identify the expression of 

38 the antigen proteins/peptides of the present invention. In detail, such methods comprise 

39 incubating a test sample with one or more nucleic acid molecules and assaying for 

40 binding of the nucleic acid molecule with components within the test sample. Such assays 

41 will typically involve arrays comprising many genes, at least one of which is a gene of the 

42 present invention and or alleles of the enzyme gene of the present invention. The figures 

43 and associated information below provide information on epitope sequence and micro- 

44 variation in strains of T. parva, that have been found in the gene encoding the antigen of 

45 the present invention. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



42 



(41860-189496) 



1 

2 Conditions for incubating a nucleic acid molecule with a test sample vary. Incubation 

3 conditions depend on the format employed in the assay, the detection methods 

4 employed, and the type and nature of the nucleic acid molecule used in the assay. One 

5 skilled in the art will recognize that any one of the commonly available hybridization, 

6 amplification or array assay formats can readily be adapted to employ the novel 

7 fragments of the T. parva genome disclosed herein. Examples of such assays can be 

8 found in Chard, T, An Introduction to Radioimmunoassay and Related Techniques, 

9 Elsevier Science Publishers, Amsterdam, The Netherlands (1986); Bullock, G. R. et al., 

10 Techniques in Immunocytochemistry, Academic Press, Orlando, Fla. Vol. 1 (1982), Vol. 2 

11 (1983), Vol. 3 (1985); Tijssen, P., Practice and Theory of Enzyme Immunoassays: 

12 Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science 

13 Publishers, Amsterdam, The Netherlands (1985). 
14 

15 The test samples of the present invention include cells, protein or membrane extracts of 

16 cells. The test sample used in the above-described method will vary based on the assay 

17 format, nature of the detection method and the tissues, cells or extracts used as the 

18 sample to be assayed. Methods for preparing nucleic acid extracts or of cells are well 

19 known in the art and can be readily be adapted in order to obtain a sample that is 

20 compatible with the system utilized. 
21 

22 In another embodiment of the present invention, kits are provided which contain the 

23 necessary reagents to carry out the assays of the present invention. 
24 

25 Specifically, the invention provides a compartmentalized kit to receive, in close 

26 confinement, one or more containers which comprises; (a) a first container comprising 

27 one of the nucleic acid molecules that can bind to a fragment of the T. parva genome 

28 disclosed herein; and (b) one or more other containers comprising one or more of the 

29 following: wash reagents, reagents capable of detecting presence of a bound nucleic 

30 acid. 
31 

32 In detail, a compartmentalized kit includes any kit in which reagents are contained in 

33 separate containers. Such containers include small glass containers, plastic containers, 

34 strips of plastic, glass or paper, or arraying material such as silica. Such containers allows 

35 one to efficiently transfer reagents from one compartment to another compartment such 

36 that the samples and reagents are not cross-contaminated, and the agents or solutions of 

37 each container can be added in a quantitative fashion from one compartment to another. 

38 Such containers will include a container which will accept the test sample, a container 

39 which contains the nucleic acid probe, containers which contain wash reagents (such as 

40 phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents 

41 used to detect the bound probe. One skilled in the art will readily recognize that the 

42 previously unidentified enzyme gene of the present invention can be routinely identified 

43 using the sequence information disclosed herein can be readily incorporated into one of 

44 the established kit formats which are well known in the art, particularly expression arrays. 
45 

46 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



43 



(41860-189496) 



1 

2 Genetically engineered host cells can be further used to produce non-human transgenic 

3 animals. A transgenic animal is preferably a mammal, for example a rodent, such as a rat 

4 or mouse, in which one or more of the cells of the animal include a transgene. A 

5 transgene is exogenous DNA which is integrated into the genome of a cell from which a 

6 transgenic animal develops and which remains in the genome of the mature animal in one 

7 or more cell types or tissues of the transgenic animal. These animals are useful for 

8 studying the function of an antigen protein and identifying and evaluating modulators of 

9 parasite development or activity. Other examples of transgenic animals include non- 
10 human primates, sheep, dogs, cows, goats, chickens, and amphibians. 

11 

12 A transgenic animal can be produced by introducing nucleic acid into the male pronuclei 

13 of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to 

14 develop in a pseudopregnant female foster animal. Any of the enzyme protein nucleotide 

15 sequences can be introduced as a transgene into the genome of a non-human animal, 

16 such as a mouse. 
17 

18 Any of the regulatory or other sequences useful in expression vectors can form part of the 

19 transgenic sequence. This includes intronic sequences and polyadenylation signals, if not 

20 already included. A tissue-specific regulatory sequence(s) can be operably linked to the 

21 transgene to direct expression of the enzyme protein to particular cells. 

22 

23 Methods for generating transgenic animals via embryo manipulation and microinjection, 

24 particularly animals such as mice, have become conventional in the art and are 

25 described, for example, in U.S. Pat. Nos. 4,736,866 and 4,870,009, both by Leder et al., 

26 U.S. Pat. No. 4,873,191 by Wagner et al. and in Hogan, B., Manipulating the Mouse 

27 Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Similar 

28 methods are used for production of other transgenic animals. A transgenic founder animal 

29 can be identified based upon the presence of the transgene in its genome and/or 

30 expression of transgenic mRNA in tissues or cells of the animals. A transgenic founder 

31 animal can then be used to breed additional animals carrying the transgene. Moreover, 

32 transgenic animals carrying a transgene can further be bred to other transgenic animals 

33 carrying other transgenes. A transgenic animal also includes animals in which the entire 

34 animal or tissues in the animal have been produced using the homologously recombinant 

35 host cells described herein. 
36 

37 In another embodiment, transgenic non-human animals can be produced which contain 

38 selected systems that allow for regulated expression of the transgene. One example of 

39 such a system is the cre/loxP recombinase system of bacteriophage P1 . For a description 

40 of the cre/loxP recombinase system, see, e.g., Lakso et al. PNAS 89:6232-6236 (1992). 

41 Another example of a recombinase system is the FLP recombinase system of S. 

42 cerevisiae (O'Gorman et al. Science 251:1351-1355 (1991). If a cre/loxP recombinase 

43 system is used to regulate expression of the transgene, animals containing transgenes 

44 encoding both the Cre recombinase and a selected protein is required. Such animals can 

45 be provided through the construction of "double" transgenic animals, e.g., by mating two 

46 transgenic animals, one containing a transgene encoding a selected protein and the other 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



44 



(41860-189496) 



1 containing a transgene encoding a recombinase. 

2 

3 Clones of the non-human transgenic animals described herein can also be produced 

4 according to the methods described in Wilmut, I. et al. Nature 385:810-813 (1997) and 

5 PCT International Publication Nos. WO97/07668 and WO97/07669. In brief, a cell, e.g., a 

6 somatic cell, from the transgenic animal can be isolated and induced to exit the growth, 

7 cycle and enter G.sub.o phase. The quiescent cell can then be fused, e.g., through the 

8 use of electrical pulses, to an enucleated oocyte from an animal of the same species from 

9 which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it 

10 develops to morula or blastocyst and then transferred to pseudopregnant female foster 

1 1 animal. The offspring born of this female foster animal will be a clone of the animal from 

12 which the cell, e.g., the somatic cell, is isolated. 
13 

14 Transgenic animals containing recombinant cells that express the peptides described 

15 herein are useful to conduct the assays described herein in an in vivo context. 

16 Accordingly, the various physiological factors that are present in vivo and that could effect 

17 substrate binding, enzyme protein activation, and signal transduction, may not be evident 

18 from in vitro cell-free or cell-based assays. Accordingly, it is useful to provide non-human 

19 transgenic animals to assay in vivo enzyme protein function, including substrate 

20 interaction, the effect of specific mutant enzyme proteins on enzyme protein function and 

21 substrate interaction, and the effect of chimeric enzyme proteins. It is also possible to 

22 assess the effect of null mutations, that is, mutations that substantially or completely 

23 eliminate one or more enzyme protein functions. 
24 

25 The T. parva antigens of the present invention have particular potential for induction of in 

26 vivo, antigen-specific CD8+ cytotoxic T cells for prophylactic immunization of cattle for the 

27 prevention of East Coast Fever disease. In addition, CTLs specific to the following 

28 metazoan parasites may also be induced by compositions identified in accordance with 

29 the methods of the present invention: Plasmodium falciparum (which causes malaria), 

30 Schistosoma mansoni (which causes schistosomiasis), and Trypanosoma cruzi (which 

31 causes Chagas' disease), Giardia lamblia, Entoemeba histolytica, Cryptospiridium spp., 

32 Leishmania spp., Brugia spp., Wuchereria spp., Onchocerca spp., Strongyloides spp., 

33 Coccidia, Haemanchus spp., Ostertagia spp., Trichomonas spp., Dirofilaria spp., 

34 Toxocara spp., Naegleria spp., Pneumocystis carinii, Ascaris spp., other Trypanosoma 

35 spp., other Schistosome spp., other Plasmodium spp., Babesia spp., Theileria spp., 

36 including but not limited to T. parva, T. lawrencei, T. annulata, T. hirci, T. ovis, T. 

37 lastoguardi, T. orientalis, T. buffeli and T. taurotragi, Babesia spp., including, but not 

38 limited to B. bigemina, B. divergens, B. major, B. bovis, B. motasi, B. ovis, B. cabelli, B. 

39 equii, B. traumani, B. canis, B. gibsoni, B. felis, and B. microfti, Adelina app., including, 

40 but not limited to A. delina, A. castana, A. picei, A. palori, and A. triboli, Anisakis and 

41 Isospora beli. 
42 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



45 



(41860-189496) 



1 Vaccination strategy 

2 

3 Cattle trials have been initiated to assess the vaccine potential of identified CTL target 

4 antigens utilizing a recombinant canarypox virus (patented by Merial Ltd) as an antigen 

5 delivery method. The first of these trials is testing one of the candidate antigens and 

6 involves use of 16 cattle. Animals were inoculated intramuscularly with 1 ml of vaccine 

7 (1x10 8 pfu of virus) and boosted similarly after 4 weeks. Following a further 4 weeks, 

8 cattle will be subjected to an LD100 challenge with T.parva sporozoites by administering 

9 subcutaneously 1 ml of diluted stabilated infective material. Animals will be monitored 

10 parasitologically and clinically over a period of 2-3 weeks to determine whether the 

11 vaccine has protected. 

12 Immunological assays will be performed following immunization and challenge to 

13 evaluate antigen-specific CTL responses and relate these to the outcome to challenge. 
14 

15 

16 All publications and patents mentioned in the above specification are herein incorporated 

17 by reference. Various modifications and variations of the described method and system of 

18 the invention will be apparent to those skilled in the art without departing from the scope 

19 and spirit of the invention. Although the invention has been described in connection with 

20 specific preferred embodiments, it should be understood that the invention as claimed 

21 should not be unduly limited to such specific embodiments. Indeed, various modifications 

22 of the above-described modes for carrying out the invention which are obvious to those 

23 skilled in the field of molecular biology or related fields are intended to be within the scope 

24 of the following outline of the invention ("claims"). 
25 

26 
27 

28 The invention is further described in the attached Annexes: 

29 ANNEX A: CTL TARGET ANTIGEN IDENTIFICATION METHOD 

30 ANNEX B: IDENTIFICATION OF A VACCINE CANDIDATE ANTIGEN RECOGNISED 

31 BY THEILERIA PARVA SPECIFIC BOVINE CYTOTOXIC T LYMPHOCYTES 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



46 



(41860489496) 



1 OUTLINE OF THE INVENTION: 

2 1) A method for the identification of parasite antigens that are targets of cytotoxic T 

3 lymphocytes, comprising the step of: co-culturing immortalized fibroblast cell lines 

4 transfected with pooled cDNA harvested from a pathogen, with clones of lines of cytotoxic T 

5 cells, generated in an animal that has been immunized, by infection and treatment with the 

6 pathogen. 

7 2) A method for a three-way matrix resolution for ^identification of a single cDNA clone from 

8 a pool of cDNAs, in high throughput procedures, comprising the steps of: 

9 a) transforming bacterial cells with DNA from a pool of about 25 to about 500 

1 0 cDNAs, that has tested positive in a routine assay; 

1 1 b) diluting the culture of transformed cells so as to yield a density of about 500-5000 

12 colonies per 150 cm2, when plated on agar-containing plates; 

13 c) picking about 100 to 500 colonies from growth cultures; 

14 d) placing about 5 to 60 pools of about 10-100 individual cultures grown from the 

15 colonies, into numbered tubes, in such a manner such that each individual bacterial 

16 culture is present in 3 of said pools, so that tubes are labelled with a number and 

17 positioned so that matrices of tubes are created so as to accommodate a multi- 

1 8 channel pipetting device; 

1 9 e) creating a corresponding matrix table is by arraying the numbers on the 

20 corresponding tubes containing the pools into an matrix table; 

21 f) testing the DNA from each of the tubes in a screening assay; and, 

22 identifying the individual positive colony by comparing the results with the array. 

23 2) The method of claim 2, wherein the screening assay causes the release of gamma interferon 

24 by CD8+ cytotoxic T cells. 

25 3) A combination of Claim 1 and Claim 2, further comprising the step of: assaying the 

26 supernatent of co-cultured cells, for the presence of soluble factors, secreted by the cytotoxic 

27 T cells. 

28 4) The method as claimed in claim 1 , wherein the pathogen is a protozoan organism. 

29 6) The method as claimed in claim 2, wherein the protozoan organism is the macroscizhont 

30 stage of an organism in the genus Theileria. 

31 3) The method as claimed in claim, 6 wherein the organism is Theileria parva. 

CONFIDENTIAL UNTIL PATENT PUBLICATION 47 



(41860-189496) 



1 4) The method as claimed in claim 4, wherein the soluble factor is a cytokine. 

2 5) The method as claimed in claim 8, wherein the cytokine in gamma interferon. 

3 6) An isolated polynucleotide represented by SEQ ID NO: 1 . (Information in Fig. 16) 

4 7) An isolated polynucleotide represented by SEQ ID NO:2. (Information in Fig. 17) 

5 8) An isolated polynucleotide represented by SEQ ID NO:3. (Information in Fig. 18) 

6 9) An isolated polynucleotide represented by SEQ ID NO:4. (Information in Fig, 19) 

7 10) An isolated polynucleotide represented by SEQ ID NO:5. (Information in Fig. 20) 

8 1 1) An isolated polypeptide represented by SEQ ID NO: 7. (Information in FIG 16.) 

9 12) An isolated polypeptide represented by SEQ ID NO: 8. (Information in FIG 17.) 

10 13) An isolated polypeptide represented by SEQ ID NO: 9. (Information in FIG 18.) 

11 14) An isolated polypeptide represented by SEQ ED NO: 10. (Information in FIG 19.) 

12 15) An isolated polypeptide represented by SEQ ID NO: 1 1 (Information in FIG 20) 

13 16) An isolated polypeptide represented by SEQ ID NO: 12 (Information in FIG. 10, as Tpl 

14 Dell, wherein said sequence stimulates cytotoxic T cells, 

15 1 7) An isolated polypeptide represented by SEQ ID NO: 13, (Information in FIG. 1 1 , as Tp 1 

16 Del2, wherein said sequence stimulates cytotoxic T cells. 

17 1 8) An isolated isolated polypeptide of the following sequence: VGYPKVKEEML. 

18 19) A method for generating polyclonal antibody in an animal wherein the immunizing antigen is 

1 9 the polypeptide represented by the sequence of one of claim 15-21. 

20 20) A method for generating monoclonal antibody, wherein the immunizing antigen is the 

2 1 polypeptide represented by the sequence of one of that of claims 15-21. 

22 21) A method comprising using of the polyclonal antibody or specific binding parts thereof, that 

23 are products of claim 23 to identify the presence of a T. parva antigen in tissue harvested 

24 from an animal. 

25 22) The use of the polyclonal antibody or specific binding parts thereof, that are products of 

26 claim 23 to quantify the expression of T. parva antigen in cells or biologic fluids. 

27 23) A diagnostic kit, for the detection of protozoan infection, using the polyclonal antibody or 

28 specific binding parts thereof, that are products of claim 23. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



(41860-189496) 



1 24) The use of the polyclonal antibody or specific binding parts thereof, that are products of 

2 claim 23 in a Western blot assay. 

3 25) The use of the polyclonal antibody or specific binding parts thereof, that are products of 

4 claim 23 in an ELIS A assay. 

5 26) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

6 claim 24 in detection of specific molecules using a flow microfluorimeter. 

7 27) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

8 claim 24 to purify antigen that binds to the antibody or its parts. 

9 28) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

1 0 claim 24 in competition assays. 

1 1 29) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

12 claim 24 to identify the presence of a T. parva antigen in tissue harvested from an animal. 

13 30) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

14 claim 24 to quantify the expression of T. parva antigen in cells or biologic fluids. 

15 3 1 ) A diagnostic kit, for the detection of protozoan infection, using the monoclonal antibody or 

16 specific binding parts thereof, that are products of claim 24. 

17 32) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

1 8 claim 24 in a Western blot assay. 

19 33) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

20 claim 24 in an ELISA assay. 

2 1 34) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

22 claim 24 in detection of specific molecules using a flow microfluorimeter. 

23 35) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

24 claim 24 to purify antigen that binds to the antibody or its parts. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



(41860-189496) 



1 36) The use of the monoclonal antibody or specific binding parts thereof, that are products of 

2 claim 24 in competition assays. 

3 37) A vector containing an isolated polynucleotide sequence comprising SEQ ID NO: 1, SEQ ID 

4 NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, or SEQ ID NO: 5. 

5 38) The vector according to claim 39, wherein said vector further comprises a polynucleotide 

6 encoding a polypeptide of interest operably linked to an isolated polynucleotide sequence 

7 comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, 

8 SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8 3 SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID 

9 NO: 11, SEQ ID NO: 12, or SEQ ID NO: 13. 

10 39) The vector according to claim 40, wherein said vector further comprises polynucleotide 

1 1 sequences encoding one or more selectable marker proteins or polypeptides, 

1 2 40) The vector of any one of claim 40, 41 , 42, or 43, wherein the vector is transfected into a cell 

13 to result in a transformed host cell. 

14 4 1) A method for the recombinant production of a polypeptide of interest comprising a vector 

15 comprising an isolated polynucleotide sequence comprising SEQ ID NO: 1, SEQ ID NO: 2, 

16 SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 

17 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, or SEQ ID NO: 13, 

1 8 operably linked to a polynucleotide encoding a polypeptide of interest and expressing said 

19 polypeptide of interest. 

20 42) The method according to claim 45, wherein said method further comprises the recovery of 

2 1 said polypeptide of interest. 

22 47) The method according to claim 45, wherein said vector further comprises polynucleotide 

23 sequences encoding one or more selectable marker proteins or polypeptides. 

24 48) The method of claim any one of claim 45, 46, or 47, further comprising transfecting the 

25 vector into host cells . 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



(41860-189496) 



1 49) The polynucleotide sequence claimed in any one of claims 10,1 1 5 12, 13 or 14, further 

2 comprising the use of such sequence as a diagnostic tool to detect nucleotide sequence in 

3 animal tissue. 

4 50) The polynucleotide sequence claimed in any one of claims 10,1 1, 12, 13 or 14, further 

5 comprising the use of such sequence as a probe of nucleic acids immobilized on a solid 

6 substrate. 

7 51) The polynucleotide sequence claimed in an one of claims 10,1 1, 12, 13 or 14, further 

8 comprising the use of such sequence to quantify the expression of mRNAs present in, or 

9 harvested from, animal tissue. 

10 52) The complimentary strand of the polynucleotide sequence claimed in any one of claims 

11 10,11, 12, 13, or 14. 

12 53) The use of the complimentary strand of claim 52 as an anti-sense polynucleotide, in either 

13 single stranded or double stranded form 

14 54) A isolated polypeptide having the following characteristics: 

15 (a) said purified protein or peptide encoded by the expression of cDNA, wherein the purified 

1 6 protein is recognized by T. parva pathogen-specific cytotoxic lymphocytes. 

17 and 

1 8 (c) said cDNA comprises; 

19 (i) nucleotide sequence that encodes a surface protein of an intracellular pathogen 

20 (ii) or a nucleotide sequence that encodes a surface protein of an intracellular pathogen that has 

21 been modified or mutated, wherein said encoded protein or peptide stimulates said cytotoxic T 

22 cells. 

23 52) A isolated polypeptide selected from the group consisting of SEQ ID NO:7, SEQ ID NO:8, 

24 SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:ll, SEQ ID NO:12 and SEQ ID NO:13. 

25 53) The isolated polypeptide according to claim 55, wherein said protein or peptide is detectable 

26 in isolates of protozoa. 

27 54) The isolated polypeptide according to claim 56, wherein said protein or peptide is detectable 

28 in isolates of Theileria. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



(41860-189496) 



1 55) The isolated polypeptide according to claim 57, wherein said protein or peptide is detectable 

2 in isolates of Theileria parva. 

3 56) An immunogenic composition comprising the polypeptide according to claim 54 and a 

4 pharmaceutically acceptable carrier. 

5 57) An immunogenic composition comprising, a cell containing an expressable recombinant 

6 nucleotide molecule, wherein the nucleotide sequence contains one or more of SEQ. ID. 

7 NO:l, SEQ. ID. NO:2, SEQ. ID. NO:3, SEQ. ID. NO:4, and SEQ. ID. NO:5. 

8 58) An immungenic composition, as in claim 60, wherein the nucleotide sequence is obtained 

9 from carrying out a PCR amplification of all or a portion of SEQ. ID. NO: 1 , SEQ. ID. NO:2, 

10 SEQ. ID. NO:3, SEQ. ID. NO:4, and SEQ. ID. NO:5. 

1 1 59) The immunogenic composition according to claim 60 or 61, wherein said composition 

12 induces cytotoxic T cells. 

13 60) A pharmaceutical composition, comprising the polypeptide of any one of claims 55, 56, 57, 

14 or 5 8 and a pharmaceutically acceptable carrier. 

15 61) A method for in vitro diagnosis of a protozoan infection which comprises contacting 

16 peripheral blood monocytes taken from an individual with the isolated polypeptide according 

17 to claim 54 or 55 under conditions suitable for binding between said polypeptide and the 

1 8 cytotoxic T lymphocytes; detecting binding between the purified protein or peptide and the 

1 9 cytotoxic T lymphocytes; and correlating the binding with the presence of the pathogentic 

20 protozoan. 

21 62) The method according to claim 64, wherein said method detects the presence of infection by 

22 a Plasmodium. 

23 63) The method according to claim 65, wherein said method detects the presence of infection by 

24 Theileria. 

25 64) The method according to claim 66, wherein said method detects the presence of infection by 

26 Theileria parva. 

27 65) The immunogenic composition of any one of claims 59, 60 or 61 , in which the antigen is 

28 administered to an animal to prevent subsequent infection by Theileria parva. 

29 66) The immunogenic composition of claim 22, in which the antigen is administered to an animal 

30 to prevent subsequent infection by Theileria parva. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



(41860-189496) 



1 

2 ABSTRACT: 
3 

4 This invention relates to DNA, protein, and peptide compositions and methods for 

5 identification of intracellular antigens of pathogens, that stimulate animal cytotoxic 

6 lymphocytes in an antigen specific manner. Such antigens are prime candidates for the 

7 development of vaccines, useful for the prevention of intracellular pathogenic diseases 

8 such as East Coast Fever. 



CONFIDENTIAL UNTIL PATENT PUBLICATION 



53 



."1 

(41860-189496) 

ANNEX A 
CTL target antigen identification 

1. Materials and methods 

2. Results 

1 . Materials and Methods 

1.1 Generation of a unidirectional cDNA expression library of Theileria 
parva schizont stage 

1.1.1 Generation of Theileria parva Muguga-infected cell lines (TpM) 

Peripheral blood mononuclear cells (PBMC) were isolated from venous blood 
and infected with T. parva (Muguga) sporozoites in accordance with published 
protocols (Goddeeris & Morrison, 1988). Infected cell lines were maintained in 
RPMI-1640 supplemented with 10% Fetaclone II (Hyclone, UK; tested for BVDV 
& Mycoplamsa spp.), 100iU/ml Penicillin, 100^g/ml Streptomycin, 50|ig/ml 
Gentamycin, 5x10~ 5 M 2-mercaptoethanol and 2mM L-Glutamine and passaged 
1/5 three times a week. 

1.1.2 Purification of schizonts 

• Harvest TpM in log growth phase, in 50 ml falcon tubes and spin down at 
1500 rpm for 10 minutes at room temperature. 

• Resuspend the cell pellet in 50 ml of plain culture medium (RPM1 1640 with 
25mMHEPES)and count. 

• Spin down again to pellet the cells. 

Annex A 
Page 1 



(41860-189496) 

Resuspend the cell pellet in the residual medium. 

Mix cell suspension with diluted antibody (anti-class I; IL-A19; dilution 1 :500 
or IL-A88, dilution 1:100), diluted in plain culture medium to yield a final 
concentration of 1x1 0 8 cells per ml. 
Incubate at on ice for 30 minutes. 

Wash of excess antibody by adding plain culture medium to 50 ml and 
centrifuging at 1000 rpm for 10 minutes at room temperature. 
Resuspend the cell pellet in 2-3 ml of plain culture medium, and add freshly 
thawed rabbit serum (Complement, previously tested for toxicity) to a final 
dilution of 1:3 to 1:5 and to a final concentration of 5x1 0 7 or 1x1 0 8 cells per ml. 
Incubate at 37 °C for 30-90 minutes either in a water bath with shaking at 15 
minutes interval until lysis is complete (check lysis by trypan blue differential 
staining using microscope). 

Spin at low speed (800 rpm) for 3 minutes to remove large cell debris. 
Transfer the schizont-rich supernatant into a fresh tube and centrifuge at 
2,500 rpm for 10 minutes at 4 °C. 

Discard the supernatant and resuspend the schizont-rich pellet in 6 ml of plain 
culture medium. 

Separate schizonts from host cell debris and nuclei by Percoll gradient 
centrifugation as described by Baumgartner et al. (Baumgartner et a/., 1999). 



Annex A 
Page 2 



(41860489496) 

1 .1 .3 Isolation of mRNA-poly(A + ) from schizonts 

• Poly(A + )RNA was isolated from frozen pellets containing approximately 
3x1 0 9 schizonts (from F100 TpM) using FastTrack 2.0 kit (Invitrogen). 
Frozen schizont pellets were thawed rapidly at room temperature then 
resuspended and lysed in 15 ml FastTrack 2.0 lysis buffer. The purification 
of poly(A + )RNA was done following the recommendations of the 
manufacturer (Invitrogen). The total amount of poly(A + )RNA isolated and 
dissolved in 20 \i\ of sterile distilled water, as estimated by ethidium- 
bromide-stained agarose gel, was 2 jig. 

• For subsequent analyses, the 20 \i\ poly(A + )RNA in a microcentrifuge tube 
was precipitated on dry-ice for 20 minutes after addition of 2 |il of 3 M 
sodium acetate buffer at pH 5.2, 50 jil of absolute ethanol. The frozen 
solution was thawed and centrifuged at 14,000 rpm for 15 minutes at 4 °C. 
The ethanol was removed and the RNA precipitate was resuspended in 1 
ml of 70% ethanol and re-centrifuged. After removing the ethanol, the 
RNA was air-dried at room temperature and re-dissolved in 6 ^il of water. It 
was then used to construct a unidirectional cDNA library using Invitrogen 
cDNA synthesis kit (Cat. No. 11917-010). 



Annex A 
Page 3 



(41860-189496) 

1 2 3 4 5 6 




Figure 1 : Ethidium bromide-stained agarose gel of RNA. Lanel , GIBCO-BRL 
RNA size markers; Iane2, bovine lymphocyte total RNA; Iane3, schizont-infected 
bovine T-lymphocytes total RNA; lane 4, schizont total RNA; lane 5, schizont 
poly(A+)-RNA; lane 6, schizont poly(A>RNA. 

1.1.4 Synthesis of cDNA 

• In a microcentrifuge tube, schizont poly(A + )RNA (2 jj,g in 6 \i\ water) was 
mixed with 2 \i\ (1 jig) of oligo(dT) Not I primer-adapter and incubated at 70 
°C for 10 minutes. 

• The mixture was spun down briefly then 4 ^l 5x first strand buffer, 2 jal 0.1 M 
DTT, 1 id 10 mM dNTPs mix and 1 \x\ [a- 32 P]dCTP (1 yCW) were added, 
mixed and incubated at 37 °C for 2 minutes. 

• After adding 4ja,l Superscript II RT, the reaction was mixed gently and 
incubated at 37 °C for 1 hour. The reaction was placed on ice. 



Annex A 
Page 4 



(41860-189496) 

One microlitre was removed and used for the calculation of the first strand 
cDNA yield. The remaining 19 \x\ was used for the second strand cDNA 
synthesis. 

To the remaining 19 \i\ first strand cDNA reaction placed on ice, 92 |J H 2 0, 30 
|iil 5x second strand buffer, 3 \xl 10 mM dNTP mix, 1 \i\ £ coli DNA ligase (10 
U/nl), 4 |xl £ coli DNA polymerase I (10 U/jJ) and 1 \i\ £ co// RNAse H (2 
U/|d) were added. 

The reaction was mixed gently, spun down briefly and incubated at 16 °C for 
2 hours. 

Two jil (10 U) T4 DNA polymerase was added to the reaction, mixed and 
incubated for additional 5 minutes at 16 °C. 

The reaction was stopped by placing the tube on ice and adding 10 ^xl 0.5 M 
EDTA. 

One hundred and fifty microlitres of phenol-chloroform solution was added to 
the cDNA reaction, and then the mixture was vortexed thoroughly for 30 
seconds and spun in a microcentrifuge at 14,000 rpm for 10 minutes at 4 °C. 
The top aqueous phase was transferred in a fresh tube and extracted with 
150 jlxI chloroform as above. 

The top aqueous phase was transferred in a fresh tube to which 75 \x\ 7.5 M 
ammonium acetate and 450 jal absolute ethanol were added and mixed. 
The tube was immediately centrifuged at 14,000 rpm for 20 minutes at room 
temperature. 



Annex A 
Page 5 



(41860-189496) 

• The supernatant was carefully removed and the cDNA pellet was gently 
overlaid with 0.5 ml of 70% ethanol. The tube was centrifuged at 14,000 rpm 
for 2 minutes at room temperature. 

• The supernatant was removed and the cDNA pellet was air-dried at room 
temperature and then redissolved in 18 jutl H2O. 

1 .1 .5 Ligation and fractionation of cDNA 

• The ligation was done using Invitrogen kit. The ligation reaction was prepared 
by adding to the tube containing the 18 of ds-cDNA the following reagents: 
10 |il 5x Adapter buffer, 10 BstX I adapter (at 1 \igl\i\ in H 2 0) 7 pi 0.1 M DTT 
and 5 nl T4 DNA ligase. The reaction was mix gently and incubated at 16 °C 
for 20 hours. 

• The ligation reaction was stopped by incubating the tube at 70 °C for 10 
minutes then chilling on ice. 

• To the 50 \i\ of ligation reaction, the following reagents were added and the 
mixture incubated at 37 °C for 90 minutes: 30 \i\ H 2 0, 10 jil 10x buffer 3 (15 
U/^il, GIBCO). 

• A Sepharose CL-2B gel filtration chromatography was prepared during Not I 
digestion reaction by washing 4 times with 0.8 ml TEN buffer (TEN buffer = 10 
mM Tris-CI, pH 7.5, 0.1 mM EDTA, 25 mM NaCL). The column is very slow 
and each wash takes about approx. 20 minutes. 

• Fifty microlitre of H2O was added to the Not I digestion (final volume: 100 nl) 
and the reaction was loaded onto the column at the center of the resin. 

Annex A 
Page 6 



(41860-189496) 

All the flow through in a single 1 .5 ml Eppendorf tube labeled tube 1 . 
Then 100 of TEN was added onto the column and the flow through 
collected in a single 1 .5 ml Eppendorf tube labeled tube 2. 
From this step, TEN was added to the resin in fractions of 100 jil and only one 
drop of the flow through was collected in each tube. Fraction collection was 
done up to tube 24. 

Each tubes containing cDNA fractions from chromatography was placed in a 
counting container (with no scintillation fluid) and counted using the tritium 
channel of a Beckman counter machine. 

Fractions representing the first 300 \i\ and fractions collected after the first 
3000 \i\ were discarded. 

The remaining were grouped into 4 fractions represented by tubes 7 to 12 
(fraction A), tubes 13 and 14 (fraction B), tubes 15 and 16 (fraction C) and 
tubes 1 7 and 18 (fraction D). The amount of cDNAs was as followed: fraction 
A, 13 ng; fraction B, 46 ng, fraction C, 95 ng; and fraction D, 82 ng. 
Five microlitre of each fraction was run on a 1% agarose gel in 0.5x TAE 
buffer at 7 V/m. The gel was then incubated in 10% TCA containing 1 % 
NaPPifor20 minutes. 

The gel was placed on a piece of Whatman 3M paper and overlaid with a pad 

of absorbent paper and a 500g weight for 10 minutes. 

The pad of absorbent paper and weight were removed and the 

gel was dried in a gel dryer under vacuum for 20 minutes. 

The dried gel was exposed to autoradiographic film at 

Annex A 
Page 7 



-80°C overnight (Figure 2). 



(41860-189496) 



A B C D 




Figure 2: Autoradiograph of electrophoresis gel of the 4 fractions (A, B, C and D) 
of -P-labelled ds-cDNA. The sizes in kilobases are indicated. 



1 .1 .6 Cloning of cDNA into pcDNA3 plasmid vector 

• Each of the 4 cDNA fractions was adjusted to 150 ^il by adding H 2 0 and 1 jjI 
(10 ng yeast t-RNA). Then 75 pi of 7.5 M NH40Ac, followed by 450 jxl of ice- 
cold absolute ethanol were added. The mixture was vortexed thoroughly and 
immediately centrifuged at room temperature for 20 min at 14,000 rpm. 

• The supernatant was removed carefully (and disposed properly), and then the 
pellet was overlaid with 0.5 ml of ice-cold 70% ethanol and centrifuged for 2 
min at 14,000 rpm. The supernatant was removed carefully (and disposed 
properly). 



Annex A 
Page 8 



(41860-189496) 

The cDNA was air-dried at room temperature and re-dissolved in H 2 0 as 
follow: fraction A in 14 fraction B in 28 pi, fraction in 70 pi and fraction D in 
56 pi. 

Bacteria E. coli DH5<x cells containing expression plasmid pcDNA3 were 
inoculated in 50 ml of 2x YT medium (tryptic soy broth 16 g/l; yeast extract, 10 
g/l; NaCI, 5.0 g/l) containing 100 pg/ml ampicillin and grown overnight at 37°C 
with vigorous shaking. 

Bacteria culture was transferred in a 50 ml falcon centrifuge tube and the cells 
were pelleted by centrifugation at 3,000 rpm in a minifuge (Heraeus Christ) for 
10 minutes at room temperature. 

The supernatant was discarded and plasmid DNA was purified from bacteria 
pellet using the QIAprepminiprep (Qiagen)) DNA purification system. Plasmid 
DNA was eluted in H 2 0. 

Twenty microgram of plasmid DNA (8 pi) was digested with 15 |jl (10 U/pl) of 
the restriction enzyme Not I (Promega) in 300 pi reaction mixture containing 
1x buffer D (Promega) at 37 °C for 3 hours. Complete digestion and 
linearization of the plasmid were confirmed by running 2 pg (30 pi) of the 
plasmid on a 0.7% agarose gel. 

The rest of the Not I reaction mix (270 pi) was mixed with 15 pi of BstX I (10 

U/pl) and 1 5 pi of water and incubated at 55 °C for 3 hours. 

Then 15 pi of the reaction was checked on a 0.7% agarose gel. 

The rest of the Not l/BstX I double digested pcDNA3 vector (285 pi) was 

dephosphorylated with 3 pi (3 U) alkaline phosphatase at 37 °C for 2 hours. 

Annex A 
Page 9 



(41860-189496) 

Digested and dephosphorylated plasmid vector was purified using CHROMA 
SPIN-1000 column (Clontech). The concentration of the vector was adjusted 
at 50 pg/ml in water. 

1 pi (50 ng) of pcDNA3 was mixed with 1 |j| (1 U) T4 DNA ligase (Promega), 1 

Ml 10x ligation buffer and 7 pi H 2 0 and incubated at 16 °C for 16 hours. 

The reaction was stopped by heating at 70 °C for 10 minutes. Then 10 pi of 

H 2 0 is added to give a final concentration of 2.5 ng/pl. 

40 \}\ of electro-competent bacteria DH5a previously prepared and tested for 

their efficiency were electroporated with 2.5 ng (1 mO of pcDNA3 at 2.49 kV/25 

MF using a cuvette of 0.2 cm electrode (BioRad) cold on ice. 

The electroporated bacteria were resuspended in 1 ml LB medium, incubated 

at 37 °C for 1 hour and then plated on LB agar plates and incubated overnight 

at 37 °C. 

Plasmid vector and ds-cDNAs were mixed in a ratio of 50 ng of vector for 
10-20 ng of cDNAs in a 20-mI ligation reaction containing 4 units of T4 DNA 
ligase (Promega). Thus the final volume of ligation of different was as fellow: 
20 pi for the 1 3 ng of fraction A, 40 m> for the 28 ng of fraction B, 100 Ml for the 
95 ng of fraction C, and 80 pi for the 82 ng of fraction D. Fraction E was a 
control ligation reaction of 40 pi containing 40 ng pcDNA3 and no ds-cDNAs. 
The ligation reactions were incubated at 4 °C for 16 hours. 
Water was added to each ligation mix to a final volume of 150 pi. Followed by 
1 pi of 10pg/plyeastt-RNA. 



Annex A 
Page 10 



(41860-189496) 

Each ligation was extracted with 150 |jl phenol-chloroform by vortexing for 30 

seconds and spinning at 14,000 rpm for 10 minutes at 4 °C. 

The top aqueous phase was re-extracted with 150 pi chloroform. 

The top aqueous phase was transferred in a fresh tube and the DNA was 

precipitated by adding 0.1 volume of 3 M NaAc, pH 5.2, and 2.5 volumes (375 

of ice-cold absolute ethanol, followed by mixing briefly and incubating 
fori 5 minutes on dry-ice. 

The tubes were thaw at room temperature and centrifuged at 14,000 rpm for 
10 minutes at 4 °C to pellet the DNA. 

The DNA pellet was washed with 1 ml of 70% ethanol, followed by 

centrifugation at 14,000 rpm for 5 minutes at 4 °C. 

The supernatant was removed and the DNA pellet was air-dried at Room 

temperature and re-dissolved in H 2 0 as follow: fraction A, 5 fj\\ fraction B, 10 

fj\\ fraction C, 25 //I; fraction D, 20 //I; and fraction E, 10 fj\. 

Frozen electrocompetent DH5a E. coli were thawed on ice and maintained on 

ice with the cuvettes. In ice-cooled Eppendorf tubes, 2.5 jj\ of DNA was mixed 

with 40 /j\ of DH5a and transferred in 0.2 cm electrode cuvette for 

electroporation at 2.49 kV/25 pF. Each electroporation was recovered with 1 

ml LB and transferred in 12 ml Sterilin tubes, followed by incubation at 37 °C 

for 1 hour on a shaker at 220 rpm. 

For fraction A, 2 electroporations were done; 4 for fraction B, 10 for fraction C, 
8 for fraction D, and only 1 for the control fraction E, Electroporations from the 
same fraction were pooled together in one tube, mixed and plated. 



Annex A 
Page 1 1 



(41860-189496) 

• For plating, 20 /j\ of each pool was transferred in a fresh tube containing 100 
jj\ of LB medium: dilution 1. 1 0 /yl of dilution 1 was transferred in another fresh 
tube containing 100 //I of LB: dilution 2; 10 jt/l of dilution 2 was transferred in 
another fresh tube containing 100 //I of LB: dilution 3. Dilutions 1, 2 and 3 
were plated onto LB agar plate containing ampicillin at 50 jug/ml for the 
titration of the library. The rest of each electroporation was plated at 2 ml per 
LB agar plate of 24x24 cm containing ampicillin at 50 /yg/ml. Plates were 
incubated overnight at 37°C. Colonies from cultures plated out at low dilutions 
were counted and the total number of colonies in each fraction calculated 
(Table 1). 



Fractions 


Number of independent 
colonies 


A 


200 


B 


12,800 


C 


355,000 


D 


298,000 


Total library 


666,000 



Table 1: Titration of cDNA library. The number of independent colonies in each 
fraction. 

Annex A 
Page 12 



(41860-189496) 

• The content of a small tube (50 jal of bacteria) was resuspended in 950 jJ of 
LB medium, mixed, diluted 10 fold several times and plated on agar plates 
containing ampicillin. 

• After incubation overnight at 37 °C, colonies formed on agar plates were 
counted and the total number of bacteria in each stabilates was determined. 



• The quality of the library was assessed as described in Table 2. 





Fraction A 


Fraction B 


Fraction C 


Fraction D 


% of colonies 
containing 
inserts 


100 


100 


95 


85 


Size of inserts 
(Kb) 


1-3 


0.4-3 


0.6-2 


0.2-1.5 


% of inserts of 
T. parva origin* 


100 


100 


100 


100 



Table 2. Analysis of randomly selected cDNA from each fraction. Plasmid DNA 
was isolated from 20 cDNA clones from each of the 4 fractions, cut with 
restriction enzymes Hind III and Xba I and analysed for presence and size of 
inserts, whose sequences were blasted against non-redundant GenBank and 
TIGR T. parva genome databases (http://www.tigr.org/tdb/e2k1/tpa1/). *An 
additional 120 cDNA were sequenced and found to be of T. parva origin. 



1.1 .7 Preparation of cDNA pools for immunoscreening with CTL 
• Pools of 50 as well as pools of 10 bacteria was prepared from fractions B and 
C followed by plasmid minipreps purification. 



Annex A 
Page 13 



(41860-189496) 

• Aliquots of frozen bacteria in small tubes containing 50 \i\ of bacteria were 
diluted and plated on agar plates to a density of 40 - 60 colonies per 4 - 6 cm 2 
and grown overnight at 37°C. 

• Colonies were scraped from the agar plate in pools of about 50 or 10 colonies 
and resuspended in 3 - 4 ml LB medium with Ampicillin. Bacteria 
suspensions were cultivated at 37°C for 3 - 4 hrs with shaking. 

• Then 0.8 ml of each culture was mixed with 0.8 ml LB medium containing 
40% glycerol and frozen as a glycerol at-80°C while the remaining was used 
for plasmid purification. Plasmid DNA was eluted in 200 y\ of water then 
transferred in a well of a 96 wells microwells plate and stored at-20°C until 
use 

• For the transfection each pool of cDNAs was adjusted at 100 ng/jxl. 

1 .1 .8 Strategy for resolving positive pools to identify cDNA coding for CTL target 
antigen 

• Competent E. coli DH5a (200 \x\) with 1 \i\ (100 ng) of DNA from the positive 
pool 

• The transformation is diluted and plated on agar plates to a density of 2000 
colonies per 150 cm 2 and grown overnight at 37°C. 

• For a positive pool of 10, only 48 individual colonies are picked and grown 
overnight in 2 ml of 2x YT medium containing ampicillin. Plasmid DNA is then 
isolated from each bacteria culture by minipreps. Plasmid DNA are eluted in 
200 |il of water and then transferred in a well of a 96 wells microwells plate 

Annex A 
Page 14 



(41860-189496) 

and stored at -20 °C. The working concentration is adjusted at 100 ng/jxl and 
stored at -20 °C until use. 

• For a positive pool of 50, a three-way matrix was used as follow: 256 
individual colonies are picked and seeded in 200 jxl of 2x YT medium 
containing ampicillin in 4 flat- bottom microwells plates. This gives 64 colonies 
per plate. 

• Bacteria were grown at 37 °C overnight. 

• From each plate containing 64 cultures, 12 pools of bacteria, each containing 
16 individual bacteria cultures, are prepared by pooling 50 |il of each 
individual bacteria culture as shown in the table 7 below. In total, 48 pools of 
16 individual bacteria are generated. Add glycerol to 20% in the each of the 
remaining cultures in Table 3 and store at -70 °C until the result of the 
screening to pick the positive colonies. 

Use of a 3-way matrix for rapid recovery of a positive cDNA clone 

Competent E. co// cells (200 were transformed with 1 |il (100 ng) of DNA 
from the positive pool of 50 cDNAs. Transformed E. coli were diluted and plated 
on agar plates to a density of 2000 colonies per 150 cm 2 and grown overnight at 
37°C. Two hundred fifty six (approximately 5x coverage of 50 cDNA per positive 
pool) individual colonies were picked and seeded in 200 |il of 2x YT medium 
containing ampicillin in 4 flat-bottom microwells plates to have 64 single bacteria 
colony cultures per plate arrayed in a 8 x 8 format and numbered from 1 to 64 as 
tabulated below (this is one of 4 plates containing 64 E. coli cultures). 



Annex A 
Page 15 



(41860-189496) 







1 


g 


17 


25 


33 


41 


49 


57 










2 


10 


18 


26 


34 


42 


50 


58 










3 


11 


19 


27 


35 


43 


51 


59 










4 


12 


20 


28 


36 


44 


52 


60 










5 


13 


21 


29 


37 


45 


53 


61 










6 


14 


22 


30 


38 


46 


54 


62 










7 


15 


23 


31 


38 


47 


55 


63 










8 


16 


24 


32 


40 


48 


56 


64 







Twelve pools of 16 individual cultures were generated from the 4 plates as 
shown in the table below. This format is high throughput in that multi-channel 
pipetting accelerates the process. The 3-way matrix is such that each individual 
bacteria culture is present in 3 pools. Pools of bacteria cultures were grown at 
37°C for 3 hours with shaking before plasmid DNA was extracted from pooled 16 
aliquots of each pool. Glycerol was added to the remaining cultures in 
microplates and stored at-70°C. Plasmid DNA was adjusted at 100 ng/jal and 
used for transfection in screening assays. A positive cDNA clone is detected in 3 
pools. For example, if colony #25 contained the target cDNA clone, pools #2, 5 
and 1 1 would be detected positive. Plasmid DNA is then prepared from the 
glycerol stock of bacteria colony #25 from the 3 pools individually and re-tested 
and sequenced. 



Annex A 
Page 16 



(41860-189496) 





P 1 


P ol 


P ol 


Pool 


Po I 


Po 1 


Po 1 


Pool 


P ol 


P ol 


P ol 


Pool 




1 


2 


3 


4 


5 


6 


7 


8 


9 


10 


11 


12 


v i 
O 


1 


17 


33 


49 


1 


2 


3 


4 


1 


5 


9 


13 


O 
3 

* 


2 


18 


34 


50 


5 


6 


7 


8 


2 


6 


10 


14 




3 


19 


35 


51 


9 


10 


11 


12 


3 


7 


11 


15 




4 


20 


36 


52 


13 


14 


15 


16 


4 


8 


12 


16 




5 


21 


37 


53 


17 


18 


19 


20 


17 


21 1 


25 


29 




6 


22 


38 


54 


21 


22 


23 


24 


18 


22 


26 


30 




7 


23 


39 


55 


25 


26 


27 


28 


19 


23 


27 


31 




8 


24 


40 


56 


29 


30 


31 


32 


20 


24 


28 


32 




9 


25 


41 


57 


33 


34 


35 


36 


33 


37 


41 


45 




10 


26 


42 


58 


37 


38 


39 


40 


34 


38 


42 


46 




11 


27 


43 


59 


41 


42 


43 


44 


35 


39 


43 


47 | 




12 


28 


44 


60 


45 


46 


47 


48 


36 


40 


44 


48 




13 


29 


45 


61 


49 


50 


51 


52 


49 


53 


57 


61 




14 


30 


46 


62 


53 


54 


55 


56 


50 


54 


58 


62 




15 


31 


47 


63 


57 


58 


59 


60 


51 


55 


59 


63 




16 


32 


48 


64 


61 


62 


63 


64 


52 


56 


60 


64 



Table 3: A 3-way matrix for rapid resolution of positive pools. In this format each 
colony is represented three times. 

• Pools of bacteria are grown at 37 °C for 3 hours with shaking before they are 
used for plasmid purification by minipreps. Plasmid DNA are eluted in 200 ^il 
of water and then transferred in a well of a 96 wells microwells plate and 
stored at -20 °C. 



Annex A 
Page 17 



(41860-189496) 

• The working concentration is adjusted at 100 ng/|il and stored at -20 °C until 
use. 

1.2 Immunisation & details of cattle 

The 7 cattle used in this invention were pure bred Bos indicus (Boran) pure bred 
Bos taurus (Friesian) or crossbreeds. Cattle were immunised by Infection and 
treatment' against the Muguga stock of T. pan/a by simultaneous inoculation of 
sporozoites and long-acting oxytetracycline at 20mg/kg BW (Radley et al 1975). 
Cryopreserved sporozoites (Stabilate # 4133) were thawed and diluted 1/20 as 
previously described. Animals were given a subcutaneous injection of 1ml of 
diluted sporozoites 2cm above the right parotid lymph node. Animals were 
monitored daily for changes in rectal temperature and from day 5 post challenge 
lymph node biopsies were taken using a 21 G needle. Giemsa stained biopsy 
smears were examined for the presence of schizont infected cells and scored on 
a scale of 1-3. Animals suffering from moderate reactions were treated with 
Buparvaquone (Butalex, Mallinckrodt Veterinary Ltd, UK). 

1.3 Generation, characterisation and maintenance of CTL 
1.3.1 Establishment of TpM 

Prior to immunisation, venous blood was collected from the eleven animals, 
PBMC were purified and infected in vitro with T. parva (Muguga) sporozoites 
(Goddeeris & Morrison, 1988). 



Annex A 
Page 18 



(41860-189496) 

1 .3.2 Generation of T. pan/a specific CD8 + CTL 

Venous blood from immunised animals was collected from 4 weeks post- 
immunisation. PBMC were prepared as described previously (Goddeeris & 
Morrison, 1988). PBMC were adjusted to 4x10 6 /ml in CTL medium (RPMI-1640 
without HEPES supplemented with 10% FBS (HyClone; tested for BVDV & 
mycoplamsa spp.), L-glutamine, 2-mercaptoethanol and antibiotics as described 
above) and 1 ml/well added to 24 well plates (Costar, Corning, NY, USA). PBMC 
were co-cultured with irradiated (50Gy) autologous T. pan/a infected cells (TpM) 
at 2 x 10 5 /well and incubated for 7 days at 37°C in a 5% C0 2 humidified 
atmosphere. Cells were harvested by aspiration and dead cells removed by 
centrifugation over Ficoll-Paque Plus (Amersham Pharmacia Biotech, Uppsala, 
Sweden). After washing in CTL medium, cells were added to 24 well plates (3x10 
6 /well) and co-cultured with irradiated PBMC (filler cells) at 1 x10 6 /well and TpM 
at 2 x10 5 /well for 7 days as before. Viable cells were harvested as described 
above, adjusted to 2 x10 6 /well and stimulated with 4 x10 5 /well irradiated TpM and 
2 x10 6 /well irradiated autologous PBMC as filler cells. 

51 Chromium Release Assay: Autologous and allogeneic TpM in log phase of 
growth were resuspended at 2x10 7 /ml cytotoxicity medium (RPMI-1640 medium 
with 5% Fetaclone II). 100//I of the target cells were mixed with 100//I (100//Ci) of 
51 Cr-sodium chromate and incubated for 1 hour at 37°C. Cells were washed 3 
times in 7ml of cytotoxicity medium by centrifugation at 1500 rpm for 7 min at RT 
and resuspended at 1X10 6 /ml. Viable cells were harvested from TpM stimulated 



Annex A 
Page 19 



(41860-189496) 

lines 7 days post-stimulation (effector cells) and resuspended in cytotoxicity 
medium at 2x10 7 /ml. Two-fold doubling dilutions of effector cells were distributed 
in duplicate (100//l/we!l) to 96-well half area (A/2) flat-bottom culture plates 
(Costar, Corning, NY, USA) resulting in a range of effector cell concentrations of 
4x1 0 6 to 2.5x1 0 5/ well. Target cells were added to each well containing effector 
ceils (50/yl/well) resulting in target cell ratios ranging from 80:1 to 5:1. In separate 
triplicate wells target cells were added to 100//I cytotoxicity medium or 1% 
Tween20 to measure spontaneous and maximal release of the label respectively. 
Plates were incubated for 4hours at 37°C in 5% CO2 humidified atmosphere. 
Cells were resuspended in wells by repeated pipetting and pelleted by 
centrifugation at 180Xg at room temperature. 75/j\ of supernatant was transferred 
from each well into sample vials (Milian, Geneva, Switzerland) and gamma 
emissions counted in a gamma counter (Micromedic MEplus, TiterTek, 
Huntsville, AL, USA). Results were calculated and expressed as percent 
cytotoxicity (= 100 x (test release - spontaneous release) / (maximum release - 
spontaneous release). 

Evidence for MHC class I restricted lysis was assessed by the capacity of 
monoclonal antibodies recognising bovine MHC class I to inhibit lysis. Cells were 
prepared for the cytotoxicity assay as described above except that target cells 
were resuspended at double the density (2x10 6 /ml) and 25//I added first to the 
plate. 25//I of either cytotoxicity medium or monoclonal antibody (mAb; IL-A88 
diluted 1/15 in cytotoxicity medium was added to target cells and incubated for 



Annex A 
Page 20 



(41860-189496) 

30min at room temperature. Serial dilutions of effector cells were then added as 
described above. 

Viable cells from TpM stimulated cultures were harvested day 7 post-infection 
and CD8 + T cells were isolated either by positive selection using flow cytometry 
(FACStar Plus, BD Biosciences, San Jose, CA, USA) or negative selection using 
Dynabeads (Dynal Biotech, Bromborough, UK). 

Positive selection: Cells were adjusted to 2x10 7 /mi in sterile monoclonal antibody 
IL-A105 (specific for bovine CD8) diluted 1/100 and incubated for 30 minutes at 
4°C. Cells were washed twice in cold culture medium and resuspended at 
2x10 7 /ml in sterile goat anti-mouse polyvalent immunoglobulins conjugated to 
FITC (Sigma). Cells were washed twice in cold medium before being run through 
a FACStar plus cell-sorter and CD8 + FITC + cells collected. 
Negative selection: Cells were adjusted to 2x10 7 /ml in sterile mAbs IL-A12 
(specific for bovine CD4) and GB21A (specific for bovine y6 TCR) diluted 1/100 
and incubated for 30 minutes at 4°C. Cells were washed twice in cold PBS and 
resuspended 1.4x10 7 /ml in sterile PBS containing washed sheep anti-mouse IgG 
Dynabeads according to the manufacturers instructions Cells and beads were 
rotated for 30 min at 4°C and Dynabead rosetted cells were collected by placing 
the sample tube in a Dynal Magnetic Particle Concentrator (MPC) for 5 min, the 
supernatant was removed, transferred to another sample tube and residual 
Dynabead rosetted cells removed by another incubation in a Dynal MPC. The 
supernatant was removed and cells washed twice in complete medium. 



Annex A 
Page 21 



(41860-189496) 

Cloning: CD8+ T cells enriched by either of the methods described above were 
adjusted to 30, 10, and 3 cell/ml and distributed into 96-well, round bottom 
culture plates containing 2 x 10 4 irradiated autologous TpM, 5 x 10 3 irradiated 
autologous PBMC and 5U/ml recombinant human IL-2 (HulL-2; Sigma, Poole, 
UK) in a final volume of 200//l/well. Plates were incubated at 37°C in humidified 
incubator containing 5% CO2 in air. Wells showing significant cell growth were 
selected for analysis of lysis of autologous TpM in an ( 111 ln) Indium oxine release 
assay. The 111 ln release assay was performed as described above for 51 Cr 
release assay except that targets cells were labelled by addition of 5//Ci 111 ln/1 x 
10 6 cells and incubation for 15 minutes at 37°C. Cells were washed five times 
with cytotoxicity medium and resuspended at 1x10 5 /ml and added 50jc/l/well to 
96-well V-bottom 96 well plates. 100//I cells from wells showing significant growth 
were transferred to 96-well V-bottom culture plates (Greiner) and centrifuged 
(180xg) for 5 min. Cells were resuspended 100//l/well in cytotoxicity medium and 
transferred to wells containing labelled target cells. Plates were centrifuged as 
described above to pellet cells before being incubated for 4 hours at 37°C in 5% 
CO2 humidified atmosphere. 

CTL populations that exhibited lytic activity on autologous infected cells and 
originated from cell dilutions that gave rise to cell growth in less than 30% of the 
wells, as they have high probability of being clones, were selected for expansion. 
The remaining cells were harvested from each well and resuspend them in 
culture medium (without HEPES) at a concentration estimated to be between 500 



Annex A 
Page 22 



(41860-189496) 

and 5000 cells/ml. 100//I of cell suspension was distributed into 96 well, round 
bottom culture plates. 100//I of autologous irradiated TpM at 5 x10 4 autologous 
irradiated TpM in medium containing 5U/ml HulL-2. Between day 14 and 21 post- 
stimulation clones and polyclonal CTL lines were subcultured in 96 well, round 
bottom culture plates by co-culturing 5000 CTL/well with 25,000 autologous 
irradiated TpM and 5U/ml HulL-2 in a final volume of 200(il/well. 

1 .4 Establishment and maintenance of bovine skin fibroblasts 

A skin biopsy was taken aseptically from the ears of cattle and placed in a 50ml 
falcon tube containing Alsever's solution. In the laboratory laminar flow hood, the 
biopsy was placed into sterile petri dishes (Sterilin) containing 1ml of 0.25% 
Trypsin-EDTA. Using a sterile scalpel blade, the sample was cut into small 
pieces and placed into a 50 ml falcon tube containing Trypsin-EDTA. The 
preparation was placed in a shaking incubator for 1-1.5 hr at 37°C with gentle 
continuous shaking. This facilitates detachment of cells. This digestion was 
stopped by addition of 1ml heat-inactivated FBS. The cell suspension was 
centrifuged for 10 min at 200xg and the cell pellet re-suspended in 6 ml of 
Dulbecco's minimum essential medium, DMEM (Gibco-BRL, Paisley, UK) 
supplemented with 10% heat-inactivated fetal bovine serum (FBS) (Hyclone, 
Logan, UT), 400 lU/ml penicillin, 300 ng/ml streptomycin and 2 mM L-glutamine. 
This cell suspension was seeded in 5ml amounts into 25-cm 2 flasks and 
incubated at 37°C in a 5% C0 2 humidified atmosphere. Cultures were examined 

Annex A 
Page 23 



(41860-189496) 

microscopically after every 4 days for growth and half the medium was replaced 
with fresh one until growing colonies were evident. Once positive colonies were 
identified they were rinsed with Ca 2+ - and Mg 2+ -free PBS/EDTA (0.02% EDTA) 
and detached by 2 min incubation at 37°C with Trypsin-EDTA solution containing 
2.5 mg/ml Trypsin and 0.2 mg/ml EDTA in HBSS (Sigma). Following a wash in 
complete DMEM containing 10% FBS the skin fibroblasts (SF) were passaged 
into 25-cm 2 tissue culture flasks (Costar). Cells were maintained in complete 
DMEM containing 10% FBS, 200 lU/ml penicillin, 100 ^ig/ml streptomycin and 2 
mM L-glutamine and passaged every 3 days at a ratio of 1:3. Cells were 
expanded in 75-cm 2 tissue culture flasks (Costar) until confluent yielding 3-4x1 0 6 
cells/flask. For consistency a liquid nitrogen cell bank was prepared with 2*10 6 
cells/freezing vial in 10% DMSO in FBS. 

1.5 Establishment and maintenance of bovine testicular endothelial cells 

Bovine testicular vein or pulmonary artery EC lines were established as 

described by Byrom and Yunker (1990) with the modifications of Mwangi et al 

(1998). Briefly, the vein was placed in wash buffer consisting of Ca 2+ - and Mg 2+ - 

free PBS supplemented with 400 Ill/ml penicillin, 400 \xg/m\ streptomycin and 

5|xg/ml fungizone. The vessel was then slit longitudinally, washed twice before 

being cut into 1-cm 2 pieces and then placed lumen side down on a drop of 

collagenase (1 mg/ml) and incubated for 1 h at 37°C. The cell suspension was 

centrifuged for 5 min at 200*g and re-suspended in 24 ml 2 * complete DMEM 

(Gibco-BRL, Paisley, UK) supplemented with 20% heat-inactivated fetal bovine 

Annex A 
Page 24 



(41860-189496) 

serum (FBS) (Hyclone, Logan, UT), 400 lU/ml penicillin, 300 |ag/ml streptomycin, 
5 mg/ml fungizone, 300 ng/ml endothelial growth supplement (Sigma, St Louis, 
MO) and 2 mM L-glutamine. One ml of the cell suspension was seeded in each 
well of a 24-well tissue culture plate (Costar, Cambridge, MA, USA). Once 
confluent, monolayers in each well were rinsed with Ca 2+ - and Mg 2+ -free 
PBS/EDTA (0.02% EDTA) and detached by 2 min incubation at 37°C with 0.25% 
trypsin/EDTA solution containing 2.5mg/ml trypsin and 0.2 mg/ml EDTA in HBSS 
(Sigma). Following a wash in DM EM containing 10% FBS the cells were 
passaged into 25-cm 2 tissue culture flasks (Costar). Cells were maintained in 
complete DMEM containing 10% FBS, 200 lU/ml penicillin, 100 ng/ml 
streptomycin, 2.5 mg/ml fungizone, 2 mM L-glutamine and passaged every 3 
days at a ratio of 1:3. Cells were expanded in 75-cm 2 tissue culture flasks 
(Costar) until confluent yielding between 3><1 0 6 and 4*10 6 cells per flask. For 
consistency a liquid nitrogen cell bank was prepared with 2><10 6 cells per freezing 
vial. Cells were raised from nitrogen into 75-cm 2 flasks and used between the 
fourth and tenth passages. 

1.6 Immortalization of bovine skin fibroblasts and endothelial cells 

Cells were immortalized with the SV40 early region gene by transfection of an 

expression plasmid psvNeo (ATCC code 37150). The product is supplied as a 

freeze dried to which 300ul of 2XYT with ampicillin was added and fully 

resuspended. The 300ul suspension was sub-cultured into 6mls and divided into 

3mls each. The cultures were incubated overnight at 37°C and then subcultured 

Annex A 
Page 25 



(41860-189496) 

again into 50mls of 2XYT with ampicillin overnight at 37°C. Maxiprep of psvNeo 
plasmid were made for use in immortalizations. Plasmid DNA for transfection 
was standardized to 2mg/ml. 

The DNA mix was prepared by mixing 5|il DNA with 495\i\ of DMEM with 
antibiotics but no serum. A working dilution of Fugene 6 transfection reagent 
(Roche) was prepared by mixing 1 5jal of Fugene with 485^1 of DMEM. Mixing 
was done using the 2ml sterile non-pyrogenic, DNAse and RNAse free 
cryopreservation tubes. The diluted Fugene was added to the diluted DNA drop 
wise with constant tapping at the end of the tube. The Fugene-DNA complex was 
allowed to form at room temperature for 30 min. 

Cells were cultured in 6-well plates (Costar) at a density of 2 x 10 5 cells per well 
and grown to confluence overnight at 37°C, 5% C0 2 in a humidified incubator. 
Culture medium was removed completely from the monolayers to be transfected. 
The Fugene-DNA complex was then added gently onto the monolayer. Plates 
were incubated at 37°C, 5% CO2 in a humidified atmosphere for 3-4 hours. The 
transfection complex was removed; fresh DMEM medium containing 10% fetal 
calf serum was added and then cultured for a further 72h. Cells from each well 
were rinsed with PBS/EDTA, detached with Trypsin/EDTA, washed and re- 
suspended in complete DMEM and sub-cultured into one T-25 flask (Costar). 
After incubation for 2-3h, normal DMEM medium was removed and replaced with 
a selection DMEM medium containing 10% FCS and 0.5jxg/ml of G418 (2.5jxg/ml 
G418 for endothelial cells) and incubated further. Upon observation of high death 
rate of cells, half the medium was replaced with fresh selection medium until 

Annex A 
Page 26 



(41860-189496) 

growing colonies were evident. This process took 3-4 weeks depending on the 
cell lines. Positive colonies were the sub-cultured into 24 well plates and then to 
T-25 flasks. Immortalization was confirmed by checking for expression of large T- 
antigen using an anti-SV40 antibody conjugated to HRP. Further expansion and 
maintenance of the cells was carried out using complete DMEM. 

1.7 Transfection of COS-7 cells and immortalised skin fibroblasts with 
schizont cDNA library 

COS-7 cells and iSF were maintained in T75 and T150 TC flasks with DMEM 
supplemented with 10% FCS, 2mM L-glutamine and antibiotics (TC medium) as 
described above. Cells are split 1:4 every 3 days. The day prior to transfection, 
cells were harvested by the removal of medium, washing in PBS and incubation 
in 0.25% Trypsin-EDTA for 5 min at 37°C. Once cells had detached TC medium 
was added and cells removed. Cells were washed by centrifugation at 1200 rpm 
for 10min and resuspended in TC medium. A viable cell count taken, density 
adjusted to 2.0x1 0 5 /ml, cells dispensed, 100//l/well, into 96 well flat-bottom TC 
plates and incubated for overnight at 37°C in a C0 2 (5%) humidified incubator. 
DNA was prepared for either single transfections of SF or double transfections 
(co-transfectton of schizont cDNA and BoLA class I cDNA) of COS cells. 6fj\ of 
schizont cDNA and 6jj\ of BoLA class I cDNA (for co-transfection) at 50ng///l in 
dH 2 0 were added to 150/yl unsupplemented DMEM in wells of a 96-well round- 
bottom plate. FuGENE 6 transfection reagent was pre-warmed to 37°C. 0.9//I or 
0.45//I FuGENE 6 was added to each well for double and single transfections 

Annex A 
Page 27 



(41860-189496) 

respectively. The well contents mixed by shaking on a Dynatech Varishaker for 1 
min and incubated at RT for 20 min. The medium from the 96 well plates 
containing adherent COS cells and SF was removed and each transfection 
complex added to triplicate wells (50)t/l/well). The cells were then incubated for 4 
hours at 37°C in a C0 2 (5%) humidified incubator. The transfection complex was 
removed and replaced with 200//l/well DM EM supplemented as described above 
and incubate for 24 or 48 hours at 37°C in a CO2 (5%) humidified incubator. 

1.8 Detection of CTL recognition of transfected SF and COS-7 cells by 
IFN^ELISpot 

24 hours post-transfection, medium was removed from wells containing 
transfected cells, cells were washed cells with PBS (200//l/well) and detached by 
the addition of 100//l/well Trypsin-EDTA as described above. Once the cells had 
detached the contents of each well were transferred to a 96 well round-bottom 
plates containing 100//l/well cold RPMI with no HEPES supplemented with 10% 
FCS (CTL medium). The cells were centrifuged at 1200 rpm for 3 min; 
supernatant removed and resuspended 50//l/well in CTL medium 
Schizont specific CTL, generated and maintained as described above, were 
harvested 7-14 days post-stimulation, transferred to polycarbonate tubes, 
pelleted at 1200 rpm for 10min and resuspended at 2x10 5 /ml in CTL medium 
supplemented with 5U/ml HulL-2 (Sigma). 



Annex A 
Page 28 



(41860-189496) 

ELISpot plates (Millipore corporation, Bedford, MA, USA) were coated 50//l/well 
with 2/yg/ml of murine anti-bovine IFN-k mAb (CC302; Serotec, UK) and 
incubated overnight at 4°C. Wells were washed twice with unsupplemented 
RPMI-1640 and blocked 20ty/l/well with RPMI-1640 supplemented with 10% FBS 
by incubating at 37°C for 2 hours. The blocking medium was removed and 
replaced with 50/y|/well CTL and 100//l/well transfected cells. As a positive 
control, irradiated TpM are serially diluted in COS cells or SF with each at a 
density of 4x10 5 /ml. and populations containing 32, 16, 8, 4, 2, 1% TpM are 
added 50/yl/well to wells containing CTL. Plates were incubated in a humidified 
incubator at 37°C for 20 hours. After incubation, the contents of wells were 
removed and wells washed four times with sterile distilled water supplemented 
with 0.05% Tween 20 per well and the plate shaken on a shaker for 30 seconds 
between washes. The process was repeated an additional four times, using PBS 
supplemented with Tween 20 (PBS-T). Wells were then incubated with 100/yl/well 
rabbit anti-bovine IFN-k antisera diluted 1/1500 in PBS-T supplemented with 
0.2% BSA (PBS/BSA) for 1 hour at room temperature. Wells were washed 4 
times with PBS-T before being incubated for 1 hour at room temperature with 
100/vl/well murine monoclonal anti-rabbit IgG conjugated to alkaline phosphatase 
(Sigma) diluted 1/2000 in PBS-T/BSA. Sigma Fast BCIP/NBT buffered substrate 
(Sigma) was by dissolving 1 tablet/1 0ml dH 2 0 and passing it through a 0.2//m 
filter. Plates were washed six times as described above with PBS-T, 100^1/well 
BCIP/NBT substrate added and plates incubated for 10minutes at room 
temperature in the dark. The substrate was then removed, wells washed with 



Annex A 
Page 29 



(41860-189496) 

copious amounts of hfeO and plates air-dried at room temperature in the dark. 
Plates were finally read on an automated ELISpot reader (AID Diagnostica, 
Strasberg, Germany). 

1.9 Bioassay for CTL activation based on rapid induction of class II MHC 
expression by constitutively negative bovine endothelial cells 

Endothelial cells were detached from confluent 75-cm 2 tissue culture flasks by 
treatment with trypsin/EDTA as described. 2.5*1 0 4 cells were seeded into each 
well of a 48-well plate in 1 ml of complete DMEM and incubated overnight. Cell- 
free test supernatants derived from either co-culture of T. pa/va-specific CD8 + T 
cell lines and SF transfected with test genes or recombinant bovine IFN-y 
(rBolFN^y, the kind gift of Ciba-Geigy, Basel, Switzerland) were dispensed in 
duplicate wells in a final volume of 160 |jJ per well. 

Following 48 h incubation at 37°C, culture supernatants were discarded and the 
monolayers washed three times in FACS medium (RPMI 1640 supplemented 
with 2% y-globulin-free horse serum (Gibco-BRL) and 0.1% sodium azide). 
Endothelial cells were labeled on ice for 30 min with 100 jal of an antibody 
cocktail comprising equal volumes of bovine class II MHC-specific monoclonal 
antibodies (mAbs) J1 1, R1 and IL-A21 (each at a 1/500 dilution of ascitic fluid). 
After three washes in FACS medium, 100 jal of FITC-conjugated goat anti-mouse 
Ig (Sigma) diluted 1/200 in FACS medium were added. After incubation at 4°C 
for 30 min and a further three washes in FACS medium, cells were detached as 

Annex A 
Page 30 



(41860-189496) 

described previously and cell surface class II MHC expression determined by 
flow cytometry, using a FACScan (Becton-Dickinson, Sunnyvale, CA, USA). The 
percentage of class II MHC-expressing cells was determined by comparison with 
unstimulated EC labeled in an identical manner. 

T. pan/a-specific CD8 + CTL lines were generated and maintained using methods 
initially described. Test supernatants were collected from 96-well flat-bottomed 
microtitre plates (Costar) 48 hours after restimulation of resting T cell lines (2x1 0 4 
per well) with 4*10 5 COS-7 cells co-transfected with the KN104 gene and test 
gene(s) or confluent autologous SF transfected with the test gene(s) in a final 
volume of 200 jliI for 24 h. These tests were set-up at least in duplicates. Where 
indicated, class I MHC was blocked using a specific antibody (IL-A88) to check 
for MHC class I restriction of the CTL lines. Additional negative control 
supernatants were derived from co-culture of CTL with untransfected 
immortalized SF or COS-7 cells. Positive control supernatants were obtained 
from co-culture of CTL with varying proportions of irradiated autologous TpM. 

1.10 Detection of CTL lysis of transfected iSF and COS-7 cells 

Autologous iSF or COS-7 cells were seeded in 6-well plates (Costar) at a density 
of 2.5x1 0 5 /well and incubated for 2 hours at 37°C to allow cells to adhere. For 
single transfections of iSF, 2^ig of test or control cDNA was added to 1ml 
unsupplemented DMEM containing 3^1 FuGENE 6 transfection reagent and 
incubated for 40min. For COS-7 cells, 2^g of test or control cDNA was added 
with 2|iig of BoLA class I cDNA to 1ml DMEM containing 6^1 FuGENE. 1ml of 

Annex A 
Page 31 



(41860-189496) 

DNA/Fugene complex was added per well of adherent COS-7 or iSF. Plates 
were incubated for 4 hours at 37°C, the transfection complex was removed and 
replaced with 2ml/well of complete DMEM and the plates incubated for a further 
20 hours at 37°C. Transfected cells were harvested by removal of medium, 
washing in PBS, detachment by Trypsin-EDTA and washing in complete DMEM. 
Transfected cells and TpM were labelled with 51 Chromium and the ability of 
schizont-specific CTL lines (day 6-8 post-stimulation) to lyse these targets was 
assessed as described above. 

1.11 Use of Exonuclease III digestion to map epitope-encoding regions 
from Tp1 

The recombinant Tp1 plasmid DNA was cut with restriction enzyme Apa I to 
generate Exonuclease III resistant 3'-protruding termini. The linearized plasmid 
DNA was then purified with phenol-chloroform before digestion with the 
restriction enzyme Xho I that will cut at the single and unique vector cloning site 
Xho I located between Not I and Apa I sites (NB: the Tp1 cDNA insert does not 
contain neither Apa I nor Xho I sites). Digestion of the DNA with Xho I generated 
an exonuclease III sensitive recessed 3'-terminus (only at the 3'side of the cDNA 
insert). The linearized plasmid DNA was purified as described previously, 100 |iig 
of DNA was digested at 37 °C with 1 unit of Exonuclease III in 20 ^il reactions at 
varying times (from 0 to 30 minutes). Reactions were stopped by 10 minutes 
incubation at 75 °C, DNA was ethanol-precipitated, washed with 70% ethanol, 
air-dried and re-dissolved in 20 |xl of 1x Mung Bean nuclease. Five units of Mung 



Annex A 
Page 32 



(41860-189496) 

Bean nuclease were added to each reaction, mixed and incubated at 30 °C for 1 
hour to remove single-stranded extensions and create ligatable blunt ends. The 
DNA was again purified with phenol-chloroform, precipitated with ethanol, 
washed with 70% ethanol, air-dried and re-dissolved in 10^1 of sterile distilled 
water. For re-ligation, 5pJ of DNA was mixed with 1^1 of 10x T4 ligation buffer, 3(il 
of water and 1jal of T4 DNA ligase, and incubated at 16 °C overnight. 
Competent E coli DH5a was transformed with 2 of the ligation and plated on 
agar plates as previously described. Plasmid DNA was isolated from single 
bacteria colonies as described and the inserts were excised by double digestion 
with BamH I and Bbs I (an isoschizomer of Bbv II). Plasmid clones with different 
cDNA inserts were selected for IFN-y ELISpot screening. 
Oligonucleotide primers were generated and used in PCR to clone small 
overlapping fragments of a 600 bp region containing the CTL epitope, into the 
mammalian expression plasmid vector pTargeT (Promega). Clones generated 
were then analysed by IFN-y ELISpot. 

1.12 Identification of Tp1 CTL epitopes with synthetic peptide libraries 

Peptide libraries (Cleaved PepSets; Mimotopes, Clayton, Australia) were 
generated for the 66 amino acid portion of Tp1 shown to contain the HD6 
restricted CTL epitope. The PepSet libraries contained every 12mer, 11mer, 
10mer and 9mer offset by 2 amino acids from the protein sequences. However, 
the peptides were prepared by truncations of the 12mers at the N-terminus and 
were supplied lyophilised with each tube containing a nominal 12mer and the 9, 

Annex A 
Page 33 



(41860-189496) 

10, 11mer truncations with the same C-terminus. Peptides were dissolved in 
400^1 50% (v/v) DNA synthesis grade acetonitrile/water (Applied Biosystems, 
Warrington, UK). To aid the dissolution, tubes were held in a sonicator water bath 
for 2 x 10 min. Peptides were aliquoted into labelled cryopreservation tubes 
(Greiner) and stored at -20°C. For screening with CTL, peptides were prepared 
at 10ng/ml in complete RPMI-1640 and 10|oJ added to triplicate wells of an 
ELISpot plate, coated, washed and blocked as described above. Autologous iSF 
or P815 cells stably expressing the BoLA class I HD6 (P815-HD6) or JSP-1 
(P815-JSP-1) were adjusted to a density of 4x10 5 /ml and 50|ul added to wells 
containing peptides. The plates were incubated at 37°C for 1 hour before CTL, 
prepared as described above for screening transfectants, were added 50|il/well. 
Plates were incubated for 20 hours at 37°C and then developed as described 
above. Based on the results of the screening with the Tp1 PepSet, individual 9, 
10 and 11mer peptides were synthesised in order to define the CTL epitopes. 
Peptides were prepared and screened using the IFN-y ELISpot as described 
above. 

Peptide-pulsed iSF and BoLA class I P815 transfectants were prepared as 
targets for 51 Chromium release assays by incubating 2x1 0 6 iSF or P815 cells 
overnight in T25 tissue culture flasks (Costar) with Tp1 peptides diluted to Vg/ml 
in complete DMEM. Cells were harvested, labelled and assayed as described 
above. 



Annex A 
Page 34 



(41860-189496) 

1.13 Detection of Tp1 pecific ex vivo CD8 + T cell responses from immun 
cattle aft r challeng with T. parva sporozoites 

Bull BV115, whose schizont specific CTL lines had been shown to recognise 
Tp1, was challenged with a lethal dose of 7. parva (Muguga) sporozoites. 
Cryopreserved sporozoites (Stabilate # 4133) were thawed and diluted 1/20 as 
previously described. Animals were challenged by subcutaneous injection of 1ml 
of diluted sporozoites 2cm above the right parotid lymph node. Animals were 
monitored daily for changes in rectal temperature and from day 5 post challenge 
lymph node biopsies were taken using a 21 G needle. Giemsa stained biopsy 
smears were examined for the presence of schizont infected cells and scored on 
a scale of 1-3. Animals were bled on day 0 and daily from day 6 to 13 and PBMC 
were isolated as described above. CD8 + T cells and CD14 + monocytes were 
purified from PBMC by MACS magnetic cell sorting according to the 
manufacturers instructions (Miltenyi Biotec, Gergisch Gladbach, Germany). CD8 + 
T cells were sorted indirectly using a monoclonal antibody specific for bovine 
CD8 (IL-A105) followed by incubation with goat anti-mouse IgG microbeads 
(Miltenyi Biotec). CD14 monocytes were sorted directly by incubation with CD14 
microbeads (Miltenyi Biotec). PBMC and CD8 + T cells were added to wells 
(2.5x10 5 /well) of coated/blocked ELISpot plates and stimulated with autologous 
TpM (2.5x1 0 4 /well) or Tp1 peptides (l^ig/ml final concentration). Purified 
monocytes were additionally added (2.5x1 0 4 /well) to wells containing peptide and 
CD8 + T cells. ELISpot plates were incubated and developed as described above. 



Annex A 
Page 35 



(41860-189496) 

In order to recall Tp1 peptide specific CTL responses, PBMC were stimulated 
with autologous TpM 14 days post-challenge as described above. Viable cells 
were harvested 7 days post-stimulation and lytic activity against TpM and Tp1 
peptide pulsed uninfected T cell blasts assessed as described above. 

2 Results of screening for CTL target antigens 

2.1 Optimisation of INF-y ELISpot for the recognition of target antigens by 
schizont specific CTL 

The ability of the IFN-y ELISpot to detect the recognition of TpM by CTL was first 
assessed using a CD8 + polyclonal CTL line from animal F100. Fourteen days 
post-stimulation, CTL were added (5000/well) to coated/blocked ELISpot wells 
containing 25,000 irradiated autologous schizont and the formation of IFN-y spots 
assessed after a 20-hour incubation. CO-culture of CTL with TpM resulted in 
significant release of IFN-y. Pre-incubating the CTL for 30 min with a mAb 
against BoLA class I completely inhibited the IFN-y response whilst mAbs against 
MHC class II or the irrelevant CD21 antigen had no effect. Significantly, there 
was almost no spontaneous release of IFN-y from CTL cultured without TpM. 
This TpM line did not constitutively express TpM and no IFN-y spots could be 
attributed to the TpM. 

In an attempt to replicate the transient transfection situation, where CTL would be 

co-cultured with COS-7 or iSF of which only a small proportion of cells would be 

Annex A 
Page 36 



(41860-189496) 

expressing the target antigen, TpM were titrated in COS-7 or iSF and co-cultured 
with CTL in IFN-y ELISpot plates. The stimulator population was fixed at an input 
of 40,000/well, with only the proportions of TpM and COS-7/SF varying. This cell 
input was adopted since it was thought to mirror the numbers of APC that would 
be co-cultured with CTL after transient transfection. Initially different CTL inputs 
were tried against TpM titrations with the aim of identifying the minimum CTL 
input required to detect significant responses to 1-3% TpM. A CTL input of 
10,000/well was determined to be optimal since it could elicit significant 
responses to less than 1 % TpM and it was practically feasible to raise such CTL 
numbers for screening experiments. 

Further titration experiments were performed with CTL clones from F100 to 
confirm that with these CTL and APC inputs the IFN-y ELISpot was meeting or 
exceeding the desired sensitivity level. With all the clones tested the IFN-y 
ELISpot could still detect recognition of target cells when they constituted only 
0.1% of the total cell population. 

The ELISpot assay worked well with little background noise and met the 
sensitivity requirements when TpM were titrated in COS-7 cells but it was 
important to determine that the assay performed as well when the TpM were 
titrated in autologous SF. Neither primary nor iSF significantly affected the 
background levels or the sensitivity of the ELISpot assay. 

In advance of the initiation of screening for CTL target antigens by the transient 
transfection of COS-7 cells and iSF, the efficiencies of COS-7 and iSF 



Annex A 
Page 37 



(41860-189496) 

transfection in 96 well TC plates was assessed using GFP as a reporter gene. 
Whilst there was considerable variation in transfection efficiencies between cell 
lines and between experiments, COS-7 consistently transfected better than iSF 
with efficiencies varying from 5-50% whereas for iSF transfection efficiency 
ranged between 0.5 - 20%. The transfection efficiency of iSF was assessed to 
be good enough to allow the presentation and identification of transfected 
schizont cDNA. 

2.2 Evaluation of IFN-y Bioassay as a complementary read-out system for 
CTL recognition of target antigens 

Bovine vascular EC were stained for surface class II MHC expression following 
culture for 48 h in the presence of media containing recombinant IFN-y. The 
sensitivity to rBolFN-y was determined and found to be between 100 and 10 
pg/ml. Comparisons were made between IFN-y bioassay and ELISpot by co- 
culturing T.pa/va-specific CTL with fixed number of autologous iSF containing 
varying proportions of target autologous TpM and skin fibroblasts. There was a 
good correlation between the two assays. Both assays detected production of 
IFN-y in co-cultures containing as low as 1% TpM. 

2.3 Identification of CTL target antigens 

2.3.1 Tp1 
a) Identification 



Annex A 
Page 38 



(41860-189496) 

A CD8 + polyclonal T cell line from Friesian bull BV115 was generated and 
maintained using an HD6 expressing 4229 TpM (4229 TpM). This CTL line, 
termed BV1 15 (4229 TpM) CD8+ polyclonal CTL line, lysed both autologous TpM 
and 4229 TpM showing that the line included schizont specific HD6 restricted 
CTL. 

This CTL line was used to screen the 1000 pools of 50 schizont cDNA (B & C 
series) following co-transfection of COS-7 cells with schizont cDNA pools and a 
pcDNA3 construct expressing the full length cDNA for HD6. The CTL line 
responded to 10 cDNA pools and the results of screening pools B1 - B200 are 
shown in Figure 3. Six of the ten putative positive pools were subjected to 
resolution and by way of illustration the results of screening the 48 resolved pools 
from the putative positive pool B162 are shown in Figure 4. The 3-way matrix 
was decoded and the single cDNA were screened, the results from screening the 
single cDNA originating from pool B162 are shown in Figure 5. Of the five single 
cDNA only three were recognised by the CTL line, all five cDNA were sequenced 
and found that the positive cDNA were identical whilst the negative cDNA were 
unrelated. It is likely that the negative cDNA were contaminated with the positive 
cDNA during propagation of the E. coli in microtitre plates since they were always 
grown in wells adjacent to wells containing the positive cDNA. 



Annex A 
Page 39 



(41860-189496) 



100 
90 
80 



^ 70 




1 25 49 73 97 121 145 169 193 

cONA pool # 



Figure 3: Screening of schizont cDNA pools (B series) with BV1 15 (4229 TpM) CD8 
polyclonal CTL. COS-7 cells co-transfected with BoLA class I HD6 cDNA and 
schizont cDNA pools were cultured with CTL and recognition assessed by IFN- 
7 ELISpot. Responses are presented as mean spot forming cell (SFC)/well. Of the 
200 cDNA pools tested, pools B42 and B162 were selected as putative positives 
and subjected to resolution. 



Annex A 
Page 40 



(41860-189496) 



O 

Li. 

CO 
C 



400 
350 
300 
250 
200 
150 
100 
50 
0 











































































I, 






1 1 I 1 1 1 1 1 1 1 1 1 I 1 1 


1 1 1 


1 1 1 1 1 1 


Mi*i 1 1 1 A i i 



1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31 33 35 37 39 41 42 45 47 

Resolved cDNA pool # 



Figure 4: Screening of resolved schizont cDNA pool B162 with BV1 15 (4229 TpM) CD8 
polyclonal CTL. 48 pools of 10 cDNA derived were constructed in a 3 way matrix and co- 
transfected with HD6 into COS-7 cells. Recognition of transfectants by CTL was 
assessed by IFN-y ELISpot. IFN-y production is presented as mean number of spot 
forming cell (SFC)/well. Of the 48 cDNA pools, 8 were positive, the 3 way matrix was 
then decoded to reveal 5 single cDNA. 



Annex A 
Page 41 



(41860-189496) 




Figure 5: Screening of single schizont cDNA with BV115 (4229 TpM) CD8 
polyclonal CTL following resolution of schizont cDNA pool B162. Five single 
cDNA decoded from the 3 way matrix were co-transfected with HD6 into COS-7 
cells. Recognition of transfectants by CTL was assessed by IFN-y ELISpot IFN- 
Y production is presented as mean number of spot forming cells (SFC)/well. Of 
the 5 single cDNA, 3 were positive, the 3 had identical sequences and were 
different from the 2 negative cDNA. 



Annex A 
Page 42 



(41860-189496) 

The response to the other resloved cDNA pools were due to this same cDNA, 
which was termed Tp1. Specific PGR primers were designed and the remaining 
putative positive pools were found to be positive for Tp1 cDNA. HD6 restriction 
recognition of Tp1 was assessed by co-transfection of Tp1 with HD6 or another 
BoLA class I cDNA, KN104 or transfection of Tp1 alone, Figure 6 shows that only 
when COS-7 cells were co-transfected with HD6 and Tp1 did the CTL secrete 
IFN-y. The ability of autologous immortalised skin fibroblasts to present Tp1 was 
assessed by transfection and compared to co-transfected COS-7 cells. Figure 7 
shows that immortalised skin fibroblasts presented Tp1 to the CTL although the 
response was less than induced by the COS-7 cells, probably due to the reduced 
transfection efficiency of skin fibroblasts. Recognition of Tp1 transfected COS-7 
cells and immortalised skin fibroblasts was also confirmed by the use of the IFN-y 
bioassay (Figure 8). 



Annex A 
Page 43 



(41860-189496) 

The ability of BV1 1 5 (4229 TpM) CD8 polyclonal CTL line to lyse Tp1 transfected 



700 
600 
500 

1 

g 400 

LL 

CO 

§ 300 
o 
2 

200 
100 
0 

Tp1 + HD6 Tp1 + KN104 Tp1 

Transfected COS-7 cell 

FIGURE 6 

targets was then assessed using a 51 Chromium release assay. COS-7 cells co- 
transfected with HD6 and Tp1 , and Tp1 transfected autologous iSF were labelled 
and used as target cells. CTL efficiently lysed 4229 TpM and Tp1 transfected 
cells (Figure 9). The lysis of Tp1 expressing targets was inhibited by the 
presence of a blocking monoclonal antibody against BoLA class I. The CTL did 
not lyse cells transfected with the irrelevant T. parva antigen PIM. 




Annex A 
Page 44 



(41860-189496) 



250 



200 



£ 150 
O 

Li. 

to 

S 100 



50 
0 



















I 1 1 1 1 



B8 B20 B42 B127 B199 B200 UnTx 
cDNA pool 

HCOS-HD6 HiSF 



Figure 7: Comparison of ability COS-7 and iSF to present target antigen containing 
schizont cDNA pools to CTL. COS-7 were co-transfected with HD6 cDNA and 
schizont cDNA pools, whilst BV115 iSF were transfected with the schizont cDNA 
pools alone. Recognition of transfectants by BV115 (4229) CD8+ polyclonal CTL 
was assessed by IFN-y ELISpot. IFN-y production is presented as mean number of 
spot forming cells (SFC)/well. Of the 7 pools tested only one, pool B42, gave a 
response above that of the untransfected cells. This pool was known to contain the 
Tp1 cDNA. The weaker response to transfected iSF is likely due to an inferior 
transfection efficiencv compared to COS-7. 



Annex A 
Page 45 




4 9 18 109 164 178 304 309 318 
cDNA 

□ HD6 BNo HD6 

Figure 8: Comparison of detection of CTL IFN-y release by ELISpot (A) and 
Bioassay (B) following recognition of Tp1 transfected COS-7 cells. 
Single cDNA isolated following resolution of positive cDNA pool B42 were 
transfected into COS-7 cells with or without HD6 cDNA. Recognition of 
transfectants by BV115 (4229) CD8+ polyclonal CTL was assessed by release of 
IFN-y using both an ELISpot and bioassay. IFN-y production as detected by 
ELISpot is presented as mean number of spot forming cells (SFC)/well and IFN-y 
bioactivity presented as % MHC class II expression. Of the 9 cDNA tested, both 
assays showed HD6 dependent recognition of 3 (#4, 178 and 309). All cDNA were 
sequenced and the positive cDNA identified as Tp1 . Annex A 

Page 46 



(41860-189496) 



70 




16:1 8:1 4:1 2:1 1:1 

E:T Ratio 



—♦-TpM 

~o-Tp1/HD6 COS 
-*-PIM/HD6 COS 
-*-Tp1/HD6 COS - class I block 
-*-Tp1 SF 
-•-PIM SF 



Figure 9: Lysis of Tp1 transfected COS-7 cells (co-transfection with BoLA class I 
HD6 cDNA) and immortalised autologous skin fibroblasts (SF) by the schizont 
specific BV115 (4229 TpM) CD8+ polyclonal CTL line. HD6 expressing 4229 TpM 
line was included as a positive control and PIM transfected COS-7 cells and iSF 
included as negative controls. MHC class I restriction was assessed by pre- 
incubating Tp1 transfected COS cells with an anti-BoLA class I mAb (MHC I block). 



Annex A 
Page 47 



(41860-189496) 

b) Mapping of the HD6 restricted Tp1 CTL epitope 

Following Exonuclease III digestion, six plasmids containing Tp1 inserts of 
differing sizes (Tp1 Dell to Del6) were co-transfected with HD6 into COS-7 cells 
and recognition of plasmid clones by BV1 15 (4229 TpM) CD8 polyclonal CTL line 
assessed by IFN-y ELISpot. Positive ELISpot responses were observed against 
clones Tp1 Dell and Del2. Both ELISpot positive and negative plasmid clones 
were sequenced and compared. Figure 10 shows a comparison of the predicted 
amino acid sequence of Tp1 with the deleted clones. The sequence comparison 
located the HD6 restricted CTL epitope within a 101 amino acid fragment of Tp1 . 
Oligonucleotide primers were generated and used in PGR to clone a 600bp 
fragment of Tp1, overlapping the epitope-encoding region (Tp1.6). IFN-y ELISpot 
confirmed CTL recognition of this portion of Tp1 and so small overlapping 
fragments (Tp1.1-Tp1.5) of the 600 bp region were cloned into the mammalian 
expression plasmid vector pTargeT. Clones were then analysed by ELISpot and 
the CTL epitope was further narrowed down to a 200 bp DNA fragment 
corresponding to 66 amino acids (Figure 1 1 ). 



Annex A 
Page 48 



io 20 30 40 bo (4t860-1894%) 

80 

Tpl ORF MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 
Tpl Dell MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 
Tpl Del2 MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 
Tpl Del 3 MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 
Tpl Del4 MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 
Tpl Del 5 MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEELKP PSALEDELKK REEESRKRME EMQKEILEKK LREGKKALEE 

Tpl Del6 MRVKKVLLYT LPWGILLAG SLIIFNFVRK RPEKEEE 

90 100 110 120 130 140 150 160 

Tpl ORF LEKREKEWD EFAKHLKKPE ERLPKIILTL DSGFPTVDPI TYTSGVYMVA VSKTTFTSDS DLVDFTHTLL GIKFLVTGVQ 
Tpl Dell LEKREKEWD EFAKHLKKPE ERLPKIILTL DSGFPTVDPI TYTSGVYMVA VSKTTFTSDS DLVDFTHTLL GIKFLVTGVQ 
Tpl Del2 LEKREKEWD EFAKHLKKPE ERLPKIILTL DSGFPTVDPI TYTSGVYMVA VSKTTFTSDS DLVDFTHTLL GIKFLVTGVQ 
Tpl Del3 LEKREKEWD EFAKHLKKPE ERLPKII . . . DSGFPTVDPI TYTSGVYMVA VSKTTFTSDS DLVDFTHTLL GIKFLVTGVQ 

Tpl Del4 LEKREKEWD EFAKHLKKPE ERLPKII 

Tpl Del 5 LEKREKEWD EFAKHLKKPE ERL 

Tpl Del6 

170 180 190 200 210 220 230 240 

Tpl ORF FGGKTYTIKP IEATMATSIA FAADPGFCYF LLIPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF NRLPKGVEIP 
Tpl Dell FGGKTYTIKP IEATMATSIA FAADPGFCYF LLIPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF NRLPKGVEIP 
Tpl Del2 FGGKTYTIKP IEATMATSIA FAADPGFCYF LLIPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF NRLPKGVEIP 

Tpl Del 3 

Tpl Del4 

Tpl Del5 

Tpl Del6 

250 260 270 280 290 300 310 320 

Tpl ORF APPGVKPEAP TPTPTTITPS VPPTIPTPIT PSAPPTTPPT GLNFNLTVQN KFMIGSQEVK LNITHEYEGV YEAHKYFIER 
Tpl Dell APPGVKPEAP TPTPTTITPS VPPTIPTPIT PSAPPTTPPT GLNFNLTVQN KFMIGSQEVK LNITHEYEGV YEAHKYFIER 

Tpl Del2 A 

Tpl Del3 

Tpl Del4 

Tpl Del5 

Tpl Del 6 

330 340 350 360 370 380 390 400 

Tpl ORF GSFTPTSFSI GDLPQTGLPV NQTVDTIWY FHRVTMGEPV GIPLIVLIFY KNQSRKYLNK GNGNWEESKA LLFREELDYL 
Tpl Dell GSFTPTSFSI GDLPQTGLPV NQTVDTIWY FHRVTMGEPV GIPLIVLIFY KNQSRKYLNK GNGNWEESKA LLFREELDYL 

Tpl Del2 

Tpl Del3 ; 

Tpl Del4 

Tpl Del5 

Tpl Del6 

410 420 430 440 450 460 470 480 

Tpl ORF DSIFNDFVTV NLSRRSDYYR NGTGTSEIEQ TLDMNVYVEP DTPCAGWTTY IHKLEEGGEG GIEKPFQIRQ LWFSKQKFDI 
Tpl Dell DSIFNDFVTV NLSRRSDYYR NGTGTSEIEQ TLDMNVYVEP DTPCAGWTTY IHKLEEGGEG GIEKPFQIRQ LWFSKQKFDI 

Tpl Del 2 

Tpl Del3 

Tpl Del4 

Tpl Del5 

Tpl Del6 

490 500 510 520 530 540 550 

Tpl ORF FPMGKVSIVN VYGKNDEPLS YAPSIFSVIR EDGIQIFYVR AYSQYLLDSS VNPQNLPQKL NTL* 

Tpl Dell FPMGKVSIVN VYGKNDEPLS YAPSIFSVIR EDGIQIFYVR AYSQYLLDSS VNPQNLPQKL NTL* 

Tpl Del2 

Tpl Del3 

Tpl Del4 

Tpl Del 5 

Tpl Del6 



Figure 10: Deduced amino acid sequences of Exonunclease III deleted Tp1 clones. 
Clones were co-transfected with HD6 cDNA and recognition by CTL determined by 
IFN-y ELISpot. Only the Tp1 orf and clones Tp1 Del! and Del2 were recognised 
implying that the HD6 restricted Tp1 epitope lay between amino acid position 141 and 
241. 



Annex A 
Page 49 



200 210 220 23( 

Tpl.l mPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF 
Tpl.2 
Tpl.3 

Tpl.4 mPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF NRLPKGVEIP 
Tpl.5 

Tpl.6 mPGPDSKP IFFKNDGDKF LRCVGYPKVK EEMLEMATKF NRLPKGVEIP 

250 260 270 280 290 

Tpl.l APPGVKPEAP TPTPTTIT 

Tpl.2 mP TPTTITPS VPPTIPTPIT PSAPPTTPPT GLNFNLTVQN 
Tpl.3 

Tpl.4 APPGVKPEAP TPTTITPSVP PTIPTPITPS APPTTPPTGL NFNLTVQNKF 

Tpl.5 mP TPTTITPSVP PTIPTPITPS APPTTPPTGL NFNLTVQNKF 

Tpl.6 APPGVKPEAP TPTTITPSVP PTIPTPITPS APPTTPPTGL NFNLTVQNKF 



300 310 320 330 340 

Tpl . 1 

Tpl.2 KFMIGSQEVK LNI THEYEGV YEAHKYFI 

Tpl.3 mGV YEAHKYFI ER GSFTPTSFSI GDLPQTGLPV 

Tpl.4 KFMIGSQEVK LNI THEYEGV YEAHKYFI ER GSFTPTSFSI GDLPQTGLPV 

Tpl.5 KFMIGSQEVK LNI THEYEGV YEAHKYFI ER GSFTPTSFSI GDLPQTGLPV 

Tpl.6 KFMIGSQEVK LNI THEYEGV YEAHKYFIER GSFTPTSFSI GDLPQTGLPV 

350 360 369 

Tpl.l 
Tpl.2 

Tpl.3 NQTVDTIWY FHRVTMGEPV GIPLIVLIF 
Tpl.4 

Tpl.5 NQTVDTIWY FHRVTMGEPV GIPLIVLIF 

Tpl.6 NQTVDTIWY FHRVTMGEPV GIPLIVLIF 



Figure 11: Deduced amino acid sequences of deleted Tp1 clones. The small m in 
bold represents the methionine added by the artificial ATG start codon by PCR. 
Constructs were co-transfected with HD6 cDNA and recognition by CTL determined 
by IFN-y ELISpot. Clones Tpl.l, 1.4 and 1.6 were recognised by CTL thus 
narrowing down the epitope containing region to 66 amino acids. 



Annex A 
Page 50 



(41860-189496) 



A cleaved PepSet library of 28 overlapping peptides each were synthesised to 
encompass the 66 amino acids encoded by the Tp1.2 insert (Mimotopes). 
Recognition of Tp1 peptides was assessed by IFN-y ELISpot, using autologous 
immortalised skin fibroblasts or murine mastocytoma P815 cells (ATCC #TIB-64, 
ATCC, Manassas, VA USA) stably expressing HD6 as antigen-presenting cells. 
Figure 12 shows the results of screening the cleaved Tp1 Pepset with BV115 
(4229 TpM) CD8 polyclonal CTL line and BV115 CTL clones. The recognition of 
peptides #10 and #11 suggested that the epitope fell within the region 
RCVGYPKVKEEMLE. All possible 9, 10 and 11mers were then synthesised for 
this sequence and screened against the polyclonal line and clones. Figure 13 
shows the responses of two BV115 CTL clones to individual Tp1 peptides. Both 
clones responded to peptide #24, the 11mer VGYPKVKEEML, suggesting that 
this was the minimal length HD6 restricted epitope of Tp1. This data was 
supported by 51 Chromium release assays that demonstrated significant CTL lysis 
of Tp1 peptide #24 pulsed iSF or P815-HD6. 



Annex A 
Page 51 



(41860-189496) 



500 

450 

400 

_ 350 

£ 300 
O 

250 

§ 200 
2 

150 
100 
50 
0 











rl 

1- 




















i I 






,j,^,^,r W| rti |r< ,, | ^ | ^ | m | J,rl,n. | n. | B ^ rm ; aM 



1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 

Tp1 peptide # 



E4229 POLY BBV115 POLY 



Figure 12: Fine mapping of the HD6 restricted Tp1 CTL epitope using synthetic 
peptide library. Twenty eight 12mer peptides overlapping by two amino acids 
encompassing the 66 amino acids encoded by the Tp1.2 insert were synthesised 
and used at a final concentration of Vg/ml to pulse BV115 iSF. Recognition of 
peptide pulsed iSF by two polyclonal CTL lines derived from BV115, one 
maintained on 4229 TpM (4229 poly) the other on autologous BV115 TpM (BV115 
poly), was assessed by IFN-y ELISpot. Significant responses were observed 
against peptides #10 and 11 that corresponded to the amino acid sequence 
RCVGYPKVKEEMLE. 



Annex A 
Page 52 



(41860-189496) 



500 

450 

400 

_ 350 

£ 300 
O 

ft 250 

§ 200 
S 

150 
100 
50 



1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 

Tp1 peptide # 

H Clone #94 ■ Clone #122 

Figure 13: Identification of the HD6 restricted Tp1 CTL epitope using synthetic 
peptides. Twenty seven peptides comprising of all the possible 9, 10 and 11mers 
from the sequence FLVGYPKVKEEMLEMA were synthesised and used at a final 
concentration of 100pg/ml to pulse P815 cells stably expressing HD6 (P815/HD6). 
Recognition of peptide pulsed P815/HD6 by two HD6 restricted BV115 CTL clones, 
#94 and #122, was assessed by IFN-y ELISpot. Significant responses were only 
observed against peptide #24, suggesting that the HD6 was the 11mer 
VGYPKVKEEML. 



Annex A 
Page 53 



(41860-189496) 



Tp1 was cloned from the Marikebuni stock of T. parva. The deduced amino acid 
sequence was compared to that of Tp1 from the Muguga stock and found to be 
95.8% identical (Figure 14). The ability of CTL to recognise the Marikabuni and 
Muguga Tp1 was assessed using the ELISpot. BV115 (4229 TpM) CD8 
polyclonal CTL line only recognised the Muguga Tp1. This is explained by a 
double amino acid substitution at the C-terminus of the predicted HD6 restricted 
epitope. 



Annex A 
Page 54 



(41860-189496) 



10 20 30 40 50 60 

TplMuguga MRVKKVLLYTLPWGILLAGSLIIFNFVRKRPEKEEEIiKPPSALEDELKKREEESRKRME 



TplMarikebuni MRVKKVLLYTLPWGILLAGSLI I FNFVRKRPEKEEELKPPSALEDELKKREEESRKRME 

10 20 30 40 50 60 

70 80 90 100 110 120 

TplMuguga EMQKEILEKKLREGKKALEELEKREKEVVDEFAKHLKKPEERLPKIILTLDSGFPTVDPI 



TplMarikebuni EMQKEILEKKLREGKKALEELEKCEKEMVDEFEKHLKKPEERLPKIILILDSGFPTVDPI 

70 80 90 100 110 120 

130 140 150 160 170 180 

TplMuguga TYTSGVYMVAVSKTTFTSDSDLVDFTHTLLGIKFLVTGVQFGGKTYTIKPI EATMATS I A 



TplMarikebuni TYTSGVYMVAVSKTTFTSDSDLVDFTHTLLGI KFLVAGVQFGGKTYTIKPIEATMATS I A 
130 140 150 160 170 180 

190 200 210 220 230 240 

TplMuguga FAADPGFCYFLLIPGPDSKPIFFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVEIP 



TplMarikebuni FAADPGFCYFLLIPGPDSKPIFFKNDGDKFLRCVGYPKVKEEIIEMATKFNRLPKGVEIP 
190 200 210 220 230 240 

250 260 270 280 290 300 

TplMuguga APPGVKPEAPTPTPTTITPSVPPTIPTPITPSAPPTTPPTGLNFNLTVQNKFMIGSQEVK 



TplMarikebuni APPGVKPEAPTPTPTTITPSVPPTIPTPITPSAPPTTPPTGLNFNLTVQNKFMVGSQEVK 
250 260 270 280 290 300 

310 320 330 340 350 360 

TplMuguga LNITHEYEGVYEAHKYFIERGSFTPTSFSIGDLPQTGLPVNQTVDTIWYFHRVTMGEPV 



TplMarikebuni LNITHEYDGVYEAHKYFIEKGRFTPTSFSIGADPQTGLPVNQTVDTIWYFHRVTMGEPV 
310 320 330 340 350 360 

370 380 390 400 410 420 

TplMuguga GIPLIVLIFYKNQSRKYLNKGNGNWEESKALLFREELDYLDSIFNDFVTVNLSRRSDYYR 



TplMarikebuni GIPLIVLVFYKNQSTKYLNKGNGNWEESKALLFREELDFLDSMFNGYVTVNLSRRSDYYR 
370 380 390 400 410 420 

430 440 450 460 470 480 

TplMuguga NGTGTSEIEQTLDMNVYVEPDTPCAGWTTYIHKLEEGGEGGIEKPFQIRQLWFSKQKFDI 



TplMarikebuni NGTGTSEIEKTLDMNVYVEPDTPCLGWTTYIHKLEEGGEGGIEKPFQIRQLWFSKQKFDI 
430 440 450 460 470 480 

490 500 510 520 530 540 

TplMuguga FPMGKVSIVNVYGKNDEPLSYAPSIFSVIREDGIQIFYVRAYSQYLLDSSVNPQNLPQKL 



TplMarikebuni FPMGKVS IVNVYGKNDEPLSYAPS IFSVIREDGI QI FYVRAYSQYLLDSSVNPQNLPQKL 
490 500 510 520 530 540 



TplMuguga NTL 
TplMarikebuni TAE 

Figure 14: Comparison of deduced amino acid sequences of Tp1 from T. parva 
Muguga and Marikebuni stocks. There was 95.8% identity in 542 aa overlap. 
Significantly there were two amino acid differences at positions 226 and 227, the C 
terminal end of the 11 amino acid HD6 restricted epitope. 

Annex A 
Page 55 



(41860-189496) 



c) Evidence that Tp1 is recognised by CD8 + T cells from an immune resolving a 
challenge infection 

Bull BV115, whose CTL had recognised Tp1, was challenged with a lethal dose 
of T. parva (Muguga) sporozoites and the response of purified CD8+ T cells to 
the HD6 restricted Tp1 eptiope or control Tp1 peptide which did not contain any 
previously identified epitopes were measured longitudinally. The animal was 
solidly resistant to challenge with no fever or detectable parasitosis. From day 8 
post-challenge, CD8+ T cells responded specifically to the Tp1 epitope and were 
sustained over the period of observation (Fig 15). The kinetics of this response is 
comparable to that previously described for schizont-speclfic CTL precursors in 
blood following challenge of immune cattle with T. parva sporozoites (McKeever 
et al 1994). Attempts were made to detect Tp1 and TpM specific lytic responses 
directly in peripheral blood post-challenge but these failed. An experiment was 
instigated to first expand schizont specific CTL numbers by a single in vitro 
stimulation with TpM and then to assess Tp1 specific lysis. Stimulated cells 
exhibited high cytotoxic activity against both TpM and T cell blasts pulsed with 
the Tp1 epitope. These data suggest that a component of the protective CD8 + T 
cell response is Tp1 -specific highlighting the potential of this antigen as a vaccine 
candidate. 



Annex A 
Page 56 




(41860-189496) 



EPITOPE 
CONTROL 



1 6 7 8 9 11 13 
Days post-challenge 



Figure 15: Tp1 specific CD8 + T cell responses of an immune bull 
following challenge with T. pa/va sporozoites. CD8 + T cells were 
purified from peripheral blood at various time-points and incubated 
with the HD6 restricted Tp1 1 1 mer synthetic epitope (epitope) or a 
Tp1 peptide which had not previously been shown to contain a 
CTL epitope (control) and responses measured by IFN-y ELISpot 
assay. 

2.3.2 Identification of other CTL target antigens 

Four other CTL target antigens, namely Tp4, Tp5, Tp7 and Tp8, have been 
identified through screening and resolution of the schizont cDNA library using 
either the ELISpot assay or bioassay. These assays utilised CTLs generated 
from five cattle representing four class I MHC genotypes. Cells transfected with 
these four antigens were recognised and lysed specifically by CTLs. Further 
assays have been performed to define the CTL epitope in Tp5. Experiments 
have been carried out which demonstrate that Tp5 is recognised by ex vivo CD8 + 
T cells obtained from an immune animal (BV050). These results are summarised 
in Table 4. 



Annex A 
Page 57 



(41860-189496) 



Antigen 


CTL 


Assay for 
ID 


Lysis 


Epitope 


Ex vivo 
response 


Tp4 


BV057 
BX063 
BX065 


ELISpot 
Bioassay 


y 

V 


Not done 


Not done 


Tp5 


BV050 


ELISpot 


/ 


/ 




Tp7 


BW012 


ELISpot 


/ 


Not done 


Not done 


Tp8 


BX063 
BX065 


ELISpot 


/ 


Not done 


Not done 



Table 4: A summary of results showing the identification of further CTL target 
antigens. 



2.4 Nucleotide and deduced amino acid sequences and putative identity 
of the 5 candidate antigens 

Following confirmation of the candidates as CTL target antigens, the individual 
cDNA were sequenced and confirmed to be of T. pan/a origin by interrogating the 
T. parva genome sequence database. BLAST searches (Altschul et al 1990) of 
DNA and protein databases were performed and homoloques of some of the 
antigens identified. SignalP (Nielsen et al 1997) and Tmpred 
(http://www.ch.embnet.org/software/TMPRED_form.html) analyses were 
conducted to predict the presence of a signal peptide and transmembrane 
domain. Figures 16-20 show the DNA and deduced protein sequences of the five 
candidate antigens. 

Nucleotide sequence of Tp1 ORF: 

aaggttaagtatagatmgtgacMatttacttacctgtttgtataaaattataaggttate^ 
ccgaWaaUgtaatatttaaagMgtacagtatATGAGGGJCAAAAAAGrm 

GTTGTCGGTATTTTACTGGCTGGATCTTTGATTATATTTMTTTCGTTAGGAAMGACCGGM 
AAAGAAGAGGAACTCAAACCTCCTTCTGCATTAGAAGATGAACTTAAAAAACGTGAAGAAGA 
AAGCCGAAAACGCATGGAAGAAATGCAAAAGGAAATTCTCGAAAAAAAGTTAAGAGAAGGTA 
AAAAAGCCTTGGAAGAACTTGAAAAACGTGAAAAAGAAGTGGTAGATGAGTTTGCAAAACAC 



Annex A 
Page 58 



(41860-189496) 



ctcaaaaaacctgaagaaagacttcctaaaattattcttacattggattccggttttccaaca 

gttgatcctattacatatacttcaggagtttatatggtagcagttagtaaaacaacttttacct 

cagattcagatcttgttgattttactcacacactgctgggcataaagtttctagttactggtg 

tacaatttggtgggaaaacatacacaattaaaccgattgaagctactatggccacttcaattg 

catttgccgctgatcctggattctgttattttctattaataccaggccctgactcgaaacca 

atattcttcaaamcgacggagatamtttttacgttgcgtagggtatccaaaggttaaa 

gaagaaatgctagaaatggctacaaaattcaatagactaccaaagggcgtggaaatacct 

gcacctccaggagtaaaaccagaggctcccacacctacaccaacgacaataactccttct 

gtacctcctactataccaacgccaataactccttcggcacctcctactacaccacctacg 

ggactaaattttaacttgacagttcagaacaaattcatgataggttcgcaagaagttaag 

ttaaatataactcacgaatacgagggtgtatacgaagctcataaatatttcattgaaagg 

ggcagctttacccctacctcattctcaatcggtgatcttccacaaacaggtcttccagta 

aatcaaacagtggatacaattgtggtatatttccatcgagtaacgatgggtgaacccgtt 

ggtatacctctmttgtgttmtcttttataaaaaccaatctagaaaatatttaaataag 

ggaaatggaaactgggaagaatctaaagctctattatttcgtgaggaacttgattactta 

gattccatttttaatgattttgtgacagtaaacctttctagacgttctgattattatcgt 

aatggaactggcacatcagaaattgagcaaacgttagatatgaatgtttacgttgaacct 

gatacaccctgtgctggatggacaacgtatatacataaattagaagaaggaggtgaagga 

ggmttgaaamccttttcamttagacaattatggtttagtaaacagaaatttgatata 

ttcccaatgggaaaagtttcaatcgttaatgtttatgggaaamcgatgaaccgctatct 

tacgctccctcmttttcagtgtaatacgcgaagatggaattcaaatattttatgttcgt 

gcttactcacagtacttgcttgattcaagtgttaatccccaaaatttacctcaaaaatta 

AACACCCTTT AGattttttttaaaaaaatcatgtaatataattgtttttgaaaaaaaaaaaaaaaaa 



Amino acid sequence of Tp1: 

MRVKKVLLYTLPWGILLAGSLIIFNFVRKRPEKEEELKPPSALEDELKKREEESR 

KRMEEMQKEILEKKLREGKKALEELEKREKEWDEFAKHLKKPEERLPKIILTLD 

SGFPTVDPITYTSGVYMVAVSKTTFTSDSDLVDFTHTLLGIKFLVTGVQFGGKTY 

TIKPIEATMATSIAFAADPGFCYFLLIPGPDSKPIFFKNDGDKFLRCVGYPKVKEE 

MLEMATKFNRLPKGVEIPAPPGVKPEAPTPTPTTITPSVPPTIPTPITPSAPPTTP 

PTGLNFNLTVQNKFMIGSQEVKLNITHEYEGVYEAHKYFIERGSFTPTSFSIGDL 

PQTGLPVNQTVDTIWYFHRVTMGEPVGIPLIVLIFYKNQSRKYLNKGNGNWEE 

SKALLFREELDYLDSIFNDFVTVNLSRRSDYYRNGTGTSEIEQTLDMNVYVEPD 

TPCAGWTTYIHKLEEGGEGGIEKPFQIRQLWFSKQKFDIFPMGKVSIVNWGKN 

DEPLSYAPSIFSVIREDGIQIFYVRAYSQYLLDSSVNPQNLPQKLNTL 

Figure 16: The nucleotide sequence of Tp1 cDNA. Tp1 cDNA contains a 
complete open reading frame (ORF) of 1632 nucleotides long shown in capital 
letters. The ATG start codon is at position 121 whereas the TAG stop codon is at 
position 1750. This ORF codes for a protein of 543 amino acids (aa). The 5'and 
3' flanked untranslated regions (UTR) are in italics. 



Annex A 
Page 59 



(41860-189496) 



Nucleotide sequence of Tp4 ORF: 

ATGAGTCATTTAATGAACCTACCAATCCTTGTATTGAAGGAAGGCACTGATA 

CATCCCAAGGCCAAGCTCAAATCATTAGTAATATCAACGCCTGTCAAGCTAT 

TGTCGATTGTGTTAAAACTACTCTAGGTCCTAGAGGAATGGACAAGTTGATA 

CATACGGAGAGAGATGTGACGATAACCAATGACGGTGCTACTGTTTTGAAAT 

TACTTGATATTACTCATCCTGCCGCTTCTGTTCTTGTTGATATCGCTAAATCA 

CAAGATGATGAAGTCGGTGATGGGACTACTTCCGTTACTGTTCTAGCAGGT 

GAGTTATTGAATGAAGCTAAGGCGTTTATATTGGATGGGATAAGTCCTCAGG 

TTATCATAAAATACTATCGTGAAGCCTGTCAAGTTGCTTTAAATCTCATTGAT 

AAAGTTGCCATTCATCTCTCCAACAAATCCTCAACTGATAAGAAAGAACTACT 

GATAAAATGTGCTGAAACTACTTTTAATTCAAAGTTATTGTCTGGTTATAAAA 

CCTTTTTTGCCAAGATGGTTGTGGAGGCAGTGGCTACTTTGGATGAGGACTT 

GGATGAGGATATGATTGGTGTTAAAAAAGTCACTGGTGGTTCCTGTGAGGA 

CTCACTCCTAGTCAAGGGTGTAGCATTCAAGAAAACTTTCAGCTACGCTGG 

GGCTGAACAACAGCCAAAGAAATTCGTCAATCCAAAGATTTTATTACTTAATT 

TGGAATTGGAACTCAAATCCGAAAAAGAAAACGCAGAAATTGTTATCAATAA 

TCCACAAGAATATCAGAAGATAATAGATGCCGAGTATAGGATAATATTTGAG 

AAGCTTGAGAATGCAGTGAAACTCGGTGCTAATGTAGTTTTATCTAAATTGC 

CMTTGGTGATTTAGCAACACAATACTTTGCAGATAAAAATGTATTTTGTGCC 

GGCCGGGTTGATGAAAATGATCTTATAAGAACGAGTAAAGCTACTGGTGCTT 

CTATTCAAACCACTCTCAATAACCTTTCAGTTGACGTCTTAGGAACTTGTGGT 

GTGTTTGAGGAAGTGCAAATTGGGTCTGAACGTTACAATATGTTCACAGATT 

GCAAGAGTGCAAAAACCTGTACAATTGTGTTGAGAGGTGGAGGTCAGCAGT 

TCATTGATGAATCTGAACGTTCACTCCATGACGCGATTATGATTGTCAGAAG 

AGCAACTAAATGTAATACTATCCTTCCCGGAGCTGGTGCCATTGAGATGTTG 

CTCTCAACTTATCTCCTCCACTATTCTCTCAACACTATTAATCCCACAGACTC 

TGTCAACCATGTTAACTGCGTTAACTCCGTAAATCATGTTAATGGAGTTACTG 

GGGTGAATAAGAGTCTGGTGGGTAAGAGGCACATAATAATGAACGGGTTTG 

CAAAG G CATTGG AGTGTATTCCAAG G AATTTAG CC ACTAATTCTGG CTACAA 

TTCAAATGATTTATTATCGATACTAAGAAATAAATACAATCAATTGGAAATAGT 

CAATGGAGAGATAAAGGTGAATAATGAGGAGAGTTGGTATGGAATAGATTG 

TTACAAGGGAAGTGTATGTAACGCATACAAGGCTTGTATTTGGGAGCCGAG 

TTTGGTGAAAAAAAACTCAATTTACTCAGCTACTGAAGCAGCTTGCCTTGTT 

CTCTCAGTTGATGAAACTGTCAAAAACCAATCCAGACAACAGTTACAAAGCG 

CACTACCACAACCCAAATAA 

Amino acid sequence of Tp4: 

MSHLMNLPILVLKEGTDTSQGQAQIISNINACQAIVDCVKTTLGPRGMDKLIHTE 

RDVTITNDGATVLKLLDITHPAASVLVDIAKSQDDEVGDGTTSVTVLAGELLNEA 

KAFILDGISPQVIIKYYREACQVALNLIDKVAIHLSNKSSTDKKELLIKCAETTFNSK 

LLSGYKTFFAKMWEAVATLDEDLDEDMIGVKKVTGGSCEDSLLVKGVAFKKTF 

SYAGAEQQPKKFVNPKILLLNLELELKSEKENAEIVINNPQEYQKIIDAEYRIIFEK 

LENAVKLGANWLSKLPIGDLATQYFADKNVFCAGRVDENDLIRTSKATGASIQT 

TLNNLSVDVLGTCGVFEEVQIGSERYNMFTDCKSAKTCTIVLRGGGQQFIDESE 

RSLHDAIMIVRRATKCNTILPGAGAIEMLLSTYLLHYSLNTINPTDSVNHVNCVNS 

VNHVNGVTGVNKSLVGKRHIIMNGFAKALECIPRNLATNSGYNSNDLLSILRNKY 



Annex A 
Page 60 



(41860-189496) 



NQLEIVNGEIKVNNEESWYGIDCYKGSVCNAYKACIWEPSLVKKNSIYSATEAA 
CLVLSVDETVKNQSRQQLQSALPQPK 



Figure 17: DNA and deduced protein sequence of Tp4. The complete ORF is 
1740 nucleotides long with an ATG start and TAA stop codons. This ORF codes 
for a protein of 579 aa. BLAST searches revealed 100% identity with the T- 
complex protein of Plasmodium and Babesia. Analysis did not predict a signal 
peptide but showed the presence of a potential transmembrane domain. 



Nucleotide sequence of Tp5 ORF: 

ATGCCGAAAAATAAAGGCAAAGGAGGAAAGAACCGGAGACGCGGTAAAAAT 

GACAATGAAGGCGAAAAAAGAGAATTAGTCTTCAAAATGGAAGATCAAGAAT 

ATGCTCMGTTTTACGTATGCTCGGTAATGGCAGACTTGAAGCCTACTGTTT 

TGACGGCACTAAACGTCTTTGCCATATTAGAGGAAAGATGAGGAAGCGAGT 

TTGGGTAAATGCCGGCGATATTATTTTGGTATCGCTTAGAGATTTCCAGGAC 

AGCAAGGCTGACGTGATCGCAAAGTACACTGCTGAGGAGGCTCGTACTCTG 

AAGGCTTACGGCGAGTTGCCTGAAGCGACCAAAATCAACGAAACTGACGTG 

TACGACGACGAGGCCGACAACTGCATTGACTTCCAGGACGTATCGTCTGAA 

TCAGAACCTGAGGATGAGTCACAAGAGGAGTCGGATTTCGATATCGATGAT 

TTATAA 

Amino acid sequence of Tp5: 

MPKNKGKGGKNRRRGKNDNEGEKRELVFKMEDQEYAQVLRMLGNGRLEAYC 

FDGTKRLCHIRGKMRKRVWVNAGDIILVSLRDFQDSKADVIAKYTAEEARTLKA 

YGELPEATKINETDVYDDEADNCIDFQDVSSESEPEDESQEESDFDIDDL 

Figure 18: DNA and deduced protein sequence of Tp5. The complete ORF is 468 
nucleotides long with an ATG start and TAA stop codons. This ORF codes for a 
protein of 1 55 aa. BLAST searches revealed that Tp5 is the 7. parva homologue 
i of the eukaryotic translation initiation factor elF-1 A (BLASTX p(n)-va\ue >10*^). 
Analysis did not predict a signal peptide or a transmembrane domain. 



Nucleotide sequence of Tp7 ORF: 

ATGACATCAAAGG ACGA GACACCTGATCAGGAGGTCTACGCTTTTAATGCT 

GA TATC TCCCAGCTTTTMGCTTGATCATCMCGCATTTTATAGTAACAAGGA 

GATTTTCCTTCGTGAACTCATTAGCAACGCTAGCGACGCACTGGAAAAAATT 

AGGTATGAGGCAATCAAGGATCCAAAGCAAATCGAGGATCAACCCGATTAC 

TATATCAGGCTGTATGCCGACAAGAACAACAACACCCTCACAATCGAAGATT 

CCGGTATTGGCATGACCAAAGCCGACCTCGTGAACAACCTCGGTACAATTG 



Annex A 
Page 61 



(41860-189496) 



CCAAATCCGGCACAAGAGCATTCATGGAGGCACTGCAAGCAGGCTCGGAC 

ATGTCAATGATCGGACAGTTTGGTGTCGGTTTCTACTCAGCATACCTGGTCG 

CAGATAAGGTGACAGTAGTGTCCAAGAACAACGCAGACGACCAGTACGTCT 

GGGAGTCAACAGCCTCAGGCCACTTTACAGTGAAGAAGGACGACTCGCAC 

GAGCCGCTCAAAAGAGGAACTAGACTAATACTGCACTTGAAGGAGGACCAA 

ACTGAGTACCTTGAGGAGAGAAGGCTGAAAGAGCTTGTTAAGAAGCACAGC 

GAGTTCATTTCATTCCCAATCTCGCTCTCAGTAGAGAAGACCCAGGAGACC 

GAGGTCACTGACGACGAGGCAGAGCTAGACGAGGACAAGAAGCCCGAGGA 

GGAGAAGCCCAAGGACGATAAGGTGGAGGACGTTACTGACGAGAAAGTGA 

CCGACGTCACTGACGAGGAGGAGAAAAAGGAGGAAAAGAAAAAGAAGAAG 

AGGAAGGTCACCAACGTAACGCGTGAGTGGGAAATGCTTAACAAGCAGAAG 

CCAATTTGGATGAGACTCCCGTCTGAAGTCACCAACGAAGAATATGCAGCG 

TTCTACAAGAACTTAACCAACGATTGGGAAGACCACTTGGCCGTGAAACACT 

TCAGCGTTGAGGGTCAGCTTGAGTTCAAAGCTCTACTGTTCGTCCCAAGAA 

GAGCGCCGTTTGACATGTTCGAGTCCCGCAAAAAGAAAAACAACATCAAGT 

TGTACGTCAGACGCGTATTTATCATGGACGACTGTGAGGAGCTCATCCCGG 

AGTGGCTTTCCTTTGTGAAGGGTGTGGTAGACTCAGAGGACCTGCCCTTGA 

ATATTTCTAGGGAAACTCTCCAGCAGAACAAGATCCTCAAGGTCATCAGGAA 

GAACTTGGTGAAAAAGTGCCTCGAGCTCTTCAATGAACTCACTGAGAAGAA 

GGAGGACTTCAAGAAGTTCTACGAGCAGTTCAGCAAGAACCTGAAGCTGGG 

AATCCACGAGGACAACGCTAATCGCTCAAAGATCGCCGAACTGTTGAGGTT 

CGAGACAACCAAGAGCGGAGACGAACTCGTGTCACTCAAGGAGTACGTTGA 

CAGGATGAAGAGTGACCAGAAGTATGTGTACTACATCACGGGAGAGTCGAA 

GCAGAGCGTAGCCTCAAGTCCTTTCCTTGAGACCCTGAGGGCTCGCGACTA 

CGAAGTCCTGTACATGACTGACCCAATTGATGAGTACGCAGTTCAGCAGAT 

CAAGGAGTTTGAAGGCAAGAAACTCAAGTGCTGTACCAAGGAGGGCCTGGA 

CCTTGATGAGGGCGAGGATGAAAAGAAGTCCTTTGAAGCGCTCAAGGAAGA 

AATGGAACCTCTTTGCAAGCACATCAAGGAAGTGCTCCACGACAAGGTGGA 

AAAGGTCGTGTGTGGAACAAGGTTTACCGACTCTCCATGCGCACTTGTCAC 

CAGCGAGTTCGGCTGGAGCGCGAACATGGAGCGTATCATGAAAGCACAAG 

CTCTCAGAGACTCGTCCATAACAAGCTACATGCTGAGCAAGAAGATCATGG 

AGATTAACCCGAGACATAGCATCATGAAGGAGCTCAAAACTAGAGCTGCAA 

ACGACAAAACAGATAAAACCGTCAAGGACCTAGTCTGGCTTCTCTACGACA 

CAGCGCTCTTAACCTCAGGGTTTAACCTCGATGAGCCCACCCAGTTTGGAA 

ACAGGATCTACAGGATGATCAAGCTCGGACTCTCATTGGACGACGAGGAAC 

ACGTAGAAGAGGACTCATCAATGCCGCCGCTGGATGAGCCCGTTGTCGACT 

CCAAAATGGAGGAAGTTGACTAA 

Amino acid sequence of Tp7: 

MTSKDETPDQEVYAFNADISQLLSLIINAFYSNKEIFLRELISNASDALEKIRYEAIK 

DPKQIEDQPDYYIRLYADKNNNTLTIEDSGIGMTKADLVNNLGTIAKSGTRAFME 

ALQAGSDMSMIGQFGVGFYSAYLVADKVTWSKNNADDQYVWESTASGHFTV 

KKDDSHEPLKRGTRLILHLKEDQTEYLEERRLKELVKKHSEFISFPISLSVEKTQE 

TEVTDDEAELDEDKKPEEEKPKDDKVEDVTDEKVTDVTDEEEKKEEKKKKKRK 

VTNVTREWEMLNKQKPIWMRLPSEVTNEEYAAFYKNLTNDWEDHLAVKHFSV 

EGQLEFKALLFVPRRAPFDMFESRKKKNNIKLYVRRVFIMDDCEELIPEWLSFVK 



Annex A 
Page 62 



(41860-189496) 



GWDSEDLPLNISRETLQQNKILKVIRKNLVKKCLELFNELTEKKEDFKKFYEQFS 

KNLKLGIHEDNANRSKIAELLRFETTKSGDELVSLKEYVDRMKSDQKYVYYITGE 

SKQSVASSPFLETLRARDYEVLYMTDPIDEYAVQQIKEFEGKKLKCCTKEGLDL 

DEGEDEKKSFEALKEEMEPLCKHIKEVLHDKVEKWCGTRFTDSPCALVTSEFG 

WSANMERIMKAQALRDSSITSYMLSKKIMEINPRHSIMKELKJRAANDKTDKTV 

KDLVWLLYDTALLTSGFNLDEPTQFGNRIYRMIKLGLSLDDEEHVEEDSSMPPL 

DEPWDSKMEEVD 



Figure 19: DNA and deduced protein sequence of Tp7. The complete ORF is 
2166 nucleotides long with an ATG start and TM stop codons. This ORF codes 
for a protein of 721 aa. BLAST searches revealed that Tp7 is the T. parva 
homologue of the heat shock protein 90 (hsp90) and showed 100% homology 
with a published partial sequence of T. parva hsp90 (accession # AAA30132). 
Analysis has predicted a potential signal peptide and a transmembrane domain. 



Nucleotide sequence of Tp8 ORF: 

ATGCTTGGAAATCATGTCATGGGATCTAATTCCCCCCACATTAAAATTTTATC 
ATCTGTTACATTCTTACATATTGCTAAAATGGAAGAAGTAGAAAACGTAAAAG 
TCGACGCCTTGGAGCGTGTTGACACTGAGTCTGTCCTTAATTATGACACTGT 

g™gaamgamccattgcgcagcagtgttgcctctttcttcaaaagatac 

agtgctgttctcgtaatattaactgccgtgctattattcacattcacttttgc 

agcaatagcattgtcatcaggcagaagcgcaatcagaaagaacagagaact 

cctgtcagtcgaatttgaaaagcttcagttcgataatttcgtgacaattaag 

ggagaaagggaagaggacttccccaagatggtagctgaagttctttacaag 

gttgcagtcgagtttgacccaaaagaagaggccttgatctacgtccagttc 

aatgacttcaacaagcaacacgacaagaagcacaacaattacaggcacaag 

aagacctcgtacaccaacttcagaaacaaccttaatgatataaacgagcaca 

acgcaaaaccaaacctgtcgtacaccaagaacatgaaccacttcggtgaca 

tatcatccaaggatttcatgaagagatacaccaagaaagtactcttgaactt 

GCCAAAAGACCACGTGTCCACCTATAACAACAACAGACCAATGTCAGTTGAT 

CTCAGAAGCCATGGTGTATTGACTCCAGTCAAGTGCCAAGAAGAAAATGAA 

CTCTCATGGCCATACTCCGTAGTAGCAGTCGCCGAGTCATTCGTTAAGAAG 

ACATCACAAAAGACCGTATCCCTCAGCGAAAAACAATTAGTAGATTGCGTTA 

CAGATAAGAAATCTGCAAACAACCCATTCTTGGGTTACAAATACCTTAAGGA 

CTTGGGTCTGTTCGAATCAGAACTCGTAGACAAATCCACAACCAAGTGCCC 

AGCATTGGAAGGTGAAAGATTCAAAGTCCCATCATACTCATACTCATATGAG 

CCAGATTTGGTGGCACTCTTGTTGAATGCAGGACCACTCACTGTACCAGTTG 

CAGTGAGCGAGGATTGGCAATTCTACGCTGATGGAACCTTGGATGTATGCG 

GTGCTGAATTGAACCACTTCTTGACCCTAGTAGGTGTCAGCTTTGACGAAAA 

AGGCAATCACTGGATACTCAAAAACTCATTCGGTGAAGGCTGGGGAAACAA 

GGGATACCTACTGTTGACTCGCAATAGCAAGGAATACAAAGATGATTGTGG 

ATTGACCTCCTTCGCAGTGTACGCAGTTTAA 



Annex A 
Page 63 



(41860-189496) 



Amino acid sequence of Tp8: 

MLGNHVMGSNSPHIKILSSVTFLHIAKMEEVENVKVDALERVDTESVLNYDTVLE 

KKPLRSSVASFFKRYSAVLVILTAVLLFTFTFAAIALSSGRSAIRKNRELLSVEFEK 

LQFDNFVTIKGEREEDFPKMVAEVLYKVAVEFDPKEEALIYVQFNDFNKQHDKK 

HNNYRHKKTSYTNFRNNLNDINEHNAKPNLSYTKNMNHFGDISSKDFMKRYTK 

KVLLNLPKDHVSTYNNNRPMSVDLRSHGVLTPVKCQEENELSWPYSWAVAES 

FVKKTSQKTVSLSEKQLVDCVTDKKSANNPFLGYKYLKDLGLFESELVDKSTTK 

CPALEGERFKVPSYSYSYEPDLVALLLNAGPLTVPVAVSEDWQFYADGTLDVC 

GAELNHFLTLVGVSFDEKGNHWILKNSFGEGWGNKGYLLLTRNSKEYKDDCGL 

TSFAVYAV 



Figure 20: DNA and deduced protein sequence of Tp8. The complete ORF is 
1323 nucleotides long with an ATG start and TAA stop codons. This ORF codes 
for a protein of 440 aa. BLAST searches revealed that Tp8 is a putative cysteine 
proteinase (BLASTX p(nj-value >10" 30 ). Analysis has predicted a potential 
transmembrane domain but no signal peptide. 



2.5 Protein Expression 



2.5.3 Construction of Plasmids 

The reading frames of T. parva candidate antigen genes were amplified by PCR 
using Taq polymerase (Promega, Madison, Wl USA 5371 1) from the original full- 
length genes in their respective plasmids in pcDNA3. Both the forward and 
reverse primers contained restriction enzyme sites (Table 1). The PCR products 
were digested with the respective restriction enzymes, and ligated into bacterial 
His-tag expression vectors, pQE30 (Qiagen, 28159 Avenue Stanford, Valencia, 
CA91355) or PET28 (Novagen, Madison, Wl 53719 USA). The plasmids were 
transformed into E. coli strain DH5a (Life Technologies, Carlsbad, CA 92008, 
USA). All the plasmids were sequenced to ensure that they harbored no 
substitutions compared to the original genes. Purified plasmids were then used to 
transform competent BL21 DE3 bacterial cells. 

Annex A 
Page 64 



(41860-189496) 



Gene 
(Plasmid) 


Forward Primer 
(Restriction Site) 


Reverse Primer 
(Restriction Site) 


Expressi 

on 
Vector 


PCR 
Product 
(s) (kb) 


Tp1 
(pcDNA3) 


GGATCCCCGGAAAAAGAAG 
AGGAACTC (BamHI) 


CTGCAGTTAAT 
TTTTGAGGTAAATTTT 
G (Pst\) 


pQE30 


1.5 


Tp4 
(pcDNA3) 


AATGTAG I I I I ATCTAAATTGCC 
A (BamH\) 


GAGGAGATAAG 
TTGAGAGCAACATC 
(Sa/I) 


pQE30 


0.4 


Tp5 
(pcDNA3) 


GGATCCGAAATGGCGAAAAATA 
AAGGCAAAGGA (BamH\) 


CTGCAGTTATAAATCA 
TCGATATCGAAATCT 
(Psfl) 


pQE30 


0.6 


Tp7 
(pcDNA3) 


GCCAAGAATTCGATGACATCAA 
AGGACGAG (gene internal SamHI 
site) 


GGCGCGGCCGCGTCA 
ACTTCCTCCAI I I IG 
(Not\,Xho\) 


pET28b 


2.0, 1.1 



Table 5: Primers, restriction sites and vectors used for the cloning of T. parva 
antigens for protein production. 

2.5.4 Expression of the proteins 

Single BL21 DE3 bacterial colonies bearing the recombinant plasmids were 
isolated and cultured in 2XYT (formula) at 37°C to an OD 6 oo of 0.6, and protein 
expression induced by addition of IPTG to a final concentration of 1mM, and 
further cultured for 4 h. The cells were harvested by centrifugation at 4000 g for 
20 min. Recombinant proteins were isolated by either the native or denaturing 
nickel-nitrilotriacetic (Ni-NTA) agarose according to the manufacturer's protocol 
(Qiagen, 28159 Avenue Stanford, Valencia, CA91355), dialyzed against PBS 
and stored at -20 °C. Purified proteins were checked on 12% SDS-PAGE gels 
and Western blot. Protein concentration was determined by the BCA Protein 
Assay reagent (PIERCE, Rockford, IL 61105, USA). Bacterial cells or purified 
proteins were applied to a 12% SDS-PAGE gel under denaturing conditions. 



Annex A 
Page 65 



(41860-189496) 



Proteins were electroblotted onto nitrocellulose sheets (Schleicher and Schull, 
Dassel, Germany). Mouse His-tag antibody (SIGMA) was used as the primary 
antibody, while anti mouse horse-raddish peroxidase conjugate (SIGMA) was 
used as the secondary antibody followed detection with 3,3-Diamnobenzidine 
and hydrogen peroxide. 

The reading frame of the segments Tp1, Tp4 and Tp5 amplified and cloned into 
the bacterial expression vectors harbored no substitutions compared to the 
original gene sequences. Recombinant protein containing His-tag were produced 
from all the constructs (Figure 21), and all can be determined by immunoblotting 
using His-tag antibody. 

Tp4 



A. 



* 8 



' mm 



4" 8 



B. 



12 12 




Coomassie Anti-His-tag 

c. 



Coomassie Anti-His-tag 



1: Crude; 2: Purified 



Tp5 



Tp5 




0) 

^ 2 o 

D O W 
Q. CO S 



SDS-PAGE WESTERN 

Coomassie Anti-His-tag 

Full-length Tp5 

Figure 21: Expression of CTL target antigens, Tp1 (A), Tp4 (B) and Tp5 (C) 
proteins. Recombinant proteins were isolated by Ni-NTA agarose and ran on 
12% SDS-PAGE gels followed by staining using coomassie blue or detection by 
Western blotting. 

Annex A 
Page 66 



(41860-189496) 



Intracellular antibody staining 




Antibody 


% of T. parva infected cells stained 


Anti-Tp1 polyclonal Ab 


78 


Anti-Tp1 monoclonal Ab 


85.96 


Anti-Tp4 polyclonal Ab 


92.52 


Anti-Tp5 polyclonal Ab 


96.04 


Control 


0.32 



Fig 22: Monoclonal and polyclonal antibodies raised against the Tp1, Tp4 and 
Tp5 proteins were used for staining of 7. parva infected cells. 



Polymorphism in CTL target antigens 

Specific Tp1 forward (IL #12588: 5'-ATG GCC ACT TCA ATT GCA TTT GCC-3' 
and reverse (IL #12589: 5'-TTA AAT GAA ATA TTT ATG AGC TTC-3') primers 
were designed and used in PCR to amplify a 430 bp region containing the BV1 15 
CTL epitope from genomic DNA of T. parva (Kakuzi521 , Kilifi KL2, Kilifi KL1 , Kilifi 
BR305, Nyairo IL02, Nyairo IL17, D409 Tp Mariakani, Buffalo7344cl, Zambia 2 
and Uganda) isolated from infected animals in different regions. PCR products 
were sequenced and their deduced amino acid sequences were compared with 
Tp1 (Figure 14). The result showed variations among the Tp1 studied. This 
strongly indicated thatTpl is polymorphic. 

Tpl MATSIAFAADPGFCYFLLI PGPDSKPIFFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVEI PAPPGVK 

Kakuz521 MATS IAFAADPGFCYFLLI PGPpjSKPI FFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVEI PAPPGVK 

NyairoILO 2 MATS IAFAADPGFCYFLLI PGPDSKPI FFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVE I PAPPGVK 

NyairoIL17 MATS IAFAADPGFCYFLLI PGPDSKPI FFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVE I PAPPGVK 

Kakuz i521 MATS IAFAADPGFCYFLLI PGPDSKPI FFKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVEIPAPPGVK 

Buffalo734 4cl MATS IAFAADPGFCYFLLI PGPDSKP I FLKNDGDKFLRCVGYPKVKEEMLEMATKFNRLPKGVE I PAPPGVK 
Kilif iKL2 MATS IAFAADPGFCYFLLI PGPDSKPI FFKNDGDKFLRCVGY PKVKEE II EMATKFNRLPKGVE I PAPPGVK 

D409TpMariakani MATSIAFAADPGICYFLLIPAP^KP I FFKNDGDKFLRCVGY PKVKEE II EMATKFNRLPKGVE I PAPPGVK 
Kilif iBR3 05 MATS IAFAADPGFCYFLLI PGPDSKP I FFKNDGDKFLRCVGYPKVKEEILEMATKFNRLPKGVE I PAPPGVK 

Kllif iKLl MATS IAFAADPGFCYFLLI PGPDSKP I FFKNDGDKFLRCVGYPKVKEE ILEMATKFNRLPKGVE I PAPPGVK 

Zambia2 . MATS IAFAADPGFCYFLLI PGPDSKP I FFKNDGDKFLRCVGY PKVKEE II EMATKFNRLPKGVE I PAPPGVK 

Uganda MATS IAFAADPGFCYFLLI PGPDSKPI FFKNDGDKFLRCVGYPKVKEEIIEMATKFNRLPKGVEI PAPPGVK 

Tpl PEAPTPTPTTITPSVPPT I PTPITPSA PPTTPPTGLNFNLTVQNKFMIGSQEVKIjNITHEYEGVYEAHKYFI 



Annex A 
Page 67 



(41860-189496) 



Kakuzi521 PE APT PT PTT I T P S VP PT IPTPITPSA P PTT P PTGLNFNLT VQNKFM I GS QE VKLN I THE YEG VYE AH KYFI 

NyairoIL02 PE APT PT PTT I T P S VPPT IPTPITPSA PPTTP PTGLNFNLT VQNKFM I G S QE VNLNI THE YEG VYE AHKYF I 

NyairoIL17 PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMIGSQEVNLNITHEYEGVYEAHKYFI 

Kakuzi521 PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMIGSQEVKLNITHEYEGVYEAHKYFI 

Buffalo7344cl PEAPTPTPTPITPSAPPT T PPTTPPKGLNFNLTLQNKFMIGSQEVKLSITHEYDGVYEAHKYFI 

KilifiKL2 PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMVGSQEVKLNITHEYDGVYEAHKYFI 

D4 0 9TpMar iakaili PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGIiNFNLTVQNKFMVGSQEVKIiNITHEYDGVYEAHKYFI 

KilifiBR305 PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMVGSQEVKLNITHEYEGVYEAHKYFI 

KilifiKLl PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMIGSPEVKLNITHEYEGVYEAHKYFI 

Zambia2 PEAPTPTPTTITP5VPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMVGSQEVKLNIPHEYDGVYEAHKYFI 

Uganda PEAPTPTPTTITPSVPPT IPTPITPSA PPTTPPTGLNFNLTVQNKFMVGSQEVKLNITHEYDGVYEAHKYFI 



FIG. 23. Tp1 multiple sequence alignments. Amino acid sequence comparison 
of a portion (containing the HD6 CTL epitope) of Tp1 generated from 7. pan/a 
isolates from different regions. Domains with variations are underlined. 



Annex A 
Page 68 




3 




5" 




7 



1 



APPLICATION DATASHEET 



Application Information 



Application Number:: 
Filing Date:: 
Application Type:: 
Subject Matter:: 



July 14, 2003 



Provisional 



Utility 



Suggested Classification:: 

Suggested Group Art Unit:: 

CD-ROM or CD-R?:: 

Number of CD Disks:: 

Number of Copies of CDs:: 

S quence Submission?:: 

Computer Readable Form 
(CFR)?:: 

Number of Copies of CFR:: 

Title:: EAST COAST FEVER VACCINE BASED ON CTL- 



Latin Name:: 

Variety Denomination Name:: 
P tition Included?:: 
P tition Type- 
Licensed US Govt. Agency:: 
Contract or Grant Numbers:: 
S cr cy Order in Parent Appl.:: 



SPECIFIC SCHIZONT ANTIGENS 



Attorney Docket Number:: 
R quest for Early Publication?:: 
R quest for Non-Publication?:: 
Suggested Drawing Figure:: 
Total Drawing Sheets:: 
Small Entity?: : 



41860-189496 



Yes 



Page 1 



Initial 07/14/03 



Applicant Information 



Applicant Authority Typ :: 
Primary Citizenship:: 
Country:: 
Status:: 



Inventor 



Given Name:: 



Full Capacity 
Evans 



Middle Name:: 



Family Name:: 



TARACHA 



Name Suffix:: 

City of Residence:: 

State or Province of Residence:: 

Country of Residence:: 

Street of Mailing Address:: 

City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Postal or Zip Code of Mailing 
Address:: 

Applicant Authority Type: : Inventor 

Primary Citizenship:: 

Country:: 

Status:: Full Capacity 

Given Name:: Jane 
Middle Name:: 

Family Name:: GLEW 
Name Suffix:: 

City of Residenc :: Nairobi 
State or Province of R sidence:: 
Country of Resid nee:: KENYA 
Street of Mailing Addr ss:: 



Page 2 



Initial 07/14/03 



City of Mailing Address:: 

State or Provinc of Mailing 
Address:: 

Country of Mailing Address- 
Postal or Zip Code of Mailing 
Address:: 

Applicant Authority Type- 
Primary Citizenship- 
Country:: 
Status:: 
Given Name:: 
Middle Name- 
Family Name- 
Name Suffix- 
City of Residence:: 
State or Province of Residence:: 
Country of Residence:: 
Street of Mailing Address- 
City of Mailing Address- 
State or Province of Mailing 
Address: : 

Country of Mailing Address- 
Postal or Zip Code of Mailing 
Address :: 

Applicant Authority Type:: 
Primary Citizenship- 
Country :: 
Status:: 
Given Nam :: 
Middl Nam :: 
Family Nam :: 



Inventor 

Full Capacity 
Simon 

GRAHAM 

Nairobi 

KENYA 



Inventor 

Full Capacity 
Duncan 

M WANG I 



Page 3 



Initial 07/14/03 



Name Suffix:: 

City of Residenc :: 

State or Province of Residence: 

Country of Residence:: 

Street of Mailing Address:: 

City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Postal or Zip Code of Mailing 
Address:: 



Nairobi 
KENYA 



Applicant Authority Type:: Inventor 
Primary Citizenship:: 

Country:: 

Status:: Full Capacity 

Given Name:: Yoshikazu 
Middle Name:: 

Family Name:: HONDA 
Name Suffix:: 

City of Residence:: Nairobi 
State or Province of Residence:: 

Country of Residence:: KENYA 
Street of Mailing Address:: 
City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Postal or Zip Code of Mailing 
Addr ss:: 



Page 4 



Initial 07/14/03 



Applicant Authority Type:: Inventor 
Primary Citizenship:: 

Country:: 

Status:: Full Capacity 

Given Name:: Roger 
Middle Name:: 

Family Name:: PELLE 
Name Suffix:: 

City of Residence: : Nairobi 
State or Province of Residence:: 

Country of Residence: : KENYA 
Street of Mailing Address:: 

City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Po tal or Zip Code of Mailing 
Address:: 



Applicant Authority Type:: Inventor 
Primary Citizenship:: 

Country:: 

Status:: Full Capacity 

Given Name:: John 
Middle Name:: 

Family Name:: TONUKARI 
Nam Suffix:: 

City of R sidenc :: Nairobi 
State or Province of Residence:: 



Page 5 



Country of R sidenc :: KENYA 
Stre t of Mailing Address:: 

City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Postal or Zip Code of Mailing 
Address:: 



Applicant Authority Type: : I nventor 

Primary Citizenship:: 

Country:: 

Status:: Full Capacity 

Given Name:: Matasuke 
Middle Name:: 

Family Name:: YAMAGE 
Name Suffix:: 

City of Residence:: 

State or Province of Residence:: 

Country of Residence:: 

Street of Mailing Address:: 

City of Mailing Address:: 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: 

Postal or Zip Code of Mailing 
Address:: 

Correspondence Information 

Corr spondenc Customer 26694 
Number:: 



Page 6 Initial 07/14/03 



Phon Number:: (202)962-4800 

Fax Number:: (202)962-8300 

E-Mail Address:: magollin@venable.com 

Representative Information 

Representative Customer 26694 
Number:: 



Domestic Priority Information 



Application:: 


Continuity Type:: 


Parent 
Application:: 


Parent Filing Date:: 




Continuation of 








Continuation of 








Continuation of 








Continuation of 







Page 7 



Initial 07/14/03 



-5 



Foreign Priority Information 



Country:: 


Application 
Number:: 


Filing Date:: 


Priority Claimed:: 



























Assignee Information 

Assignee Name:: International Livestock Research Institute 

Street of Mailing Address:: 

City of Mailing Address:: Nairobi 

State or Province of Mailing 
Address:: 

Country of Mailing Address:: KENYA 

Postal or Zip Code of Mailing 
Address:: 



Page 8 



Initial 07/14/03